1
|
Soloviova K, Via CS. Sex differences in donor T cell targeting of host splenocyte subpopulations in acute and chronic murine graft-vs.-host disease: implications for lupus-like autoimmunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.595177. [PMID: 38915570 PMCID: PMC11195085 DOI: 10.1101/2024.06.07.595177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
This study sought to compare in vivo sex differences in either a Th1-dominant CTL response or a Tfh-mediated lupus-like antibody response using the parent-into F1 murine model of acute or chronic GVHD respectively. In acute GVHD we observed no significant sex differences in the hierarchy of donor CD8 CTL elimination of splenocyte subsets. B cells were the most sensitive to elimination in both sexes; however, the male response was significantly stronger. Sex differences in chronic GVHD were more widespread; females exhibited significantly greater numbers of total splenocytes and host CD4 Tfh cells, B cells and CD8 T cells consistent with reports of greater female autoantibody production in this model. The more potent male CTL response in acute GVHD conflicts with reports of greater female CTL responses following infections or vaccines and may reflect the absence of exogenous innate immune stimuli in this model.
Collapse
Affiliation(s)
- Kateryna Soloviova
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda MD 20814
| | - Charles S Via
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda MD 20814
| |
Collapse
|
2
|
Singh RP, Bischoff DS, Hahn BH. CD8 + T regulatory cells in lupus. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2021; 2:147-156. [PMID: 35880241 PMCID: PMC9242525 DOI: 10.2478/rir-2021-0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/23/2021] [Indexed: 04/11/2023]
Abstract
T regulatory cells (Tregs) have a key role in the maintenance of immune homeostasis and the regulation of immune tolerance by preventing the inflammation and suppressing the autoimmune responses. Numerical and functional deficits of these cells have been reported in systemic lupus erythematosus (SLE) patients and mouse models of SLE, where their imbalance and dysregulated activities have been reported to significantly influence the disease pathogenesis, progression and outcomes. Most studies in SLE have focused on CD4+ Tregs and it has become clear that a critical role in the control of immune tolerance after the breakdown of self-tolerance is provided by CD8+ Tregs. Here we review the role, cellular and molecular phenotypes, and mechanisms of action of CD8+ Tregs in SLE, including ways to induce these cells for immunotherapeutic modulation in SLE.
Collapse
Affiliation(s)
- Ram P. Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, USA
| | - David S. Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bevra H. Hahn
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
3
|
Datta SK. Harnessing Tolerogenic Histone Peptide Epitopes From Nucleosomes for Selective Down-Regulation of Pathogenic Autoimmune Response in Lupus (Past, Present, and Future). Front Immunol 2021; 12:629807. [PMID: 33936042 PMCID: PMC8080879 DOI: 10.3389/fimmu.2021.629807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Autoantigen-directed tolerance can be induced by certain nucleosomal histone peptide epitope/s in nanomolar dosage leading to sustained remission of disease in mice with spontaneous SLE. By contrast, lupus is accelerated by administration of intact (whole) histones, or whole nucleosomes in microparticles from apoptotic cells, or by post-translationally acetylated histone-peptides. Low-dose therapy with the histone-peptide epitopes simultaneously induces TGFβ and inhibits IL-6 production by DC in vivo, especially pDC, which then induce CD4+CD25+ Treg and CD8+ Treg cells that suppress pathogenic autoimmune response. Both types of induced Treg cells are FoxP3+ and act by producing TGFβ at close cell-to-cell range. No anaphylactic adverse reactions, or generalized immunosuppression have been detected in mice injected with the peptides, because the epitopes are derived from evolutionarily conserved histones in the chromatin; and the peptides are expressed in the thymus during ontogeny, and their native sequences have not been altered. The peptide-induced Treg cells can block severe lupus on adoptive transfer reducing inflammatory cell reaction and infiltration in the kidney. In Humans, similar potent Treg cells are generated by the histone peptide epitopes in vitro in lupus patients’ PBMC, inhibiting anti-dsDNA autoantibody and interferon production. Furthermore, the same types of Treg cells are generated in lupus patients who are in very long-term remission (2-8 years) after undergoing autologous hematopoietic stem cell transplantation. These Treg cells are not found in lupus patients treated conventionally into clinical remission (SLEDAI of 0); and consequently they still harbor pathogenic autoimmune cells, causing subclinical damage. Although antigen-specific therapy with pinpoint accuracy is suitable for straight-forward organ-specific autoimmune diseases, Systemic Lupus is much more complex. The histone peptide epitopes have unique tolerogenic properties for inhibiting Innate immune cells (DC), T cells and B cell populations that are both antigen-specifically and cross-reactively involved in the pathogenic autoimmune response in lupus. The histone peptide tolerance is a natural and non-toxic therapy suitable for treating early lupus, and also maintaining lupus patients after toxic drug therapy. The experimental steps, challenges and possible solutions for successful therapy with these peptide epitopes are discussed in this highly focused review on Systemic Lupus.
Collapse
Affiliation(s)
- Syamal K Datta
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
4
|
Sisto M, Ribatti D, Lisi S. Organ Fibrosis and Autoimmunity: The Role of Inflammation in TGFβ-Dependent EMT. Biomolecules 2021; 11:biom11020310. [PMID: 33670735 PMCID: PMC7922523 DOI: 10.3390/biom11020310] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
Recent advances in our understanding of the molecular pathways that control the link of inflammation with organ fibrosis and autoimmune diseases point to the epithelial to mesenchymal transition (EMT) as the common association in the progression of these diseases characterized by an intense inflammatory response. EMT, a process in which epithelial cells are gradually transformed to mesenchymal cells, is a major contributor to the pathogenesis of fibrosis. Importantly, the chronic inflammatory microenvironment has emerged as a decisive factor in the induction of pathological EMT. Transforming growth factor-β (TGF-β), a multifunctional cytokine, plays a crucial role in the induction of fibrosis, often associated with chronic phases of inflammatory diseases, contributing to marked fibrotic changes that severely impair normal tissue architecture and function. The understanding of molecular mechanisms underlying EMT-dependent fibrosis has both a basic and a translational relevance, since it may be useful to design therapies aimed at counteracting organ deterioration and failure. To this end, we reviewed the recent literature to better elucidate the molecular response to inflammatory/fibrogenic signals in autoimmune diseases in order to further the specific regulation of EMT-dependent fibrosis in more targeted therapies.
Collapse
|
5
|
Muhammad Yusoff F, Wong KK, Mohd Redzwan N. Th1, Th2, and Th17 cytokines in systemic lupus erythematosus. Autoimmunity 2019; 53:8-20. [PMID: 31771364 DOI: 10.1080/08916934.2019.1693545] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the breakdown of immune tolerance leading to excessive inflammation and tissue damage. Imbalance in the levels of cytokines represents one of the multifactorial causes of SLE pathogenesis and it contributes to disease severity. Deregulated levels of T helper type 1 (Th1), type 2 (Th2), and type 17 (Th17) cytokines have been associated with autoimmune inflammation. Growing evidence has shown deregulated levels of Th1, Th2, and Th17 cytokines in SLE patients compared to healthy controls associated with disease activity and severity. In this review, we describe and discuss the levels of Th1, Th2, and Th17 cytokines in SLE patients, and clinical trials involving Th1, Th2, and Th17 cytokines in SLE patients. In particular, with the exception of IL-2, IL-4, and TGF-β1, the levels of Th1, Th2, and Th17 cytokines are increased in SLE patients associated with disease severity. Current phase II or III studies involve therapeutic antibodies targeting IFN-α and type I IFN receptor, while low-dose IL-2 therapy is assessed in phase II clinical trials.
Collapse
Affiliation(s)
- Farhana Muhammad Yusoff
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu, Malaysia
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu, Malaysia
| | - Norhanani Mohd Redzwan
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu, Malaysia
| |
Collapse
|
6
|
Wu Q, Yang Q, Sun H. Collagen triple helix repeat containing-1: a novel biomarker associated with disease activity in Systemic lupus erythematosus. Lupus 2018; 27:2076-2085. [PMID: 30336754 DOI: 10.1177/0961203318804877] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The objective of this article is to investigate whether the aberrant expression of collagen triple helix repeat containing-1 (CTHRC1) from patients with systemic lupus erythematosus (SLE) could contribute to the pathogenesis of lupus. METHODS We divided SLE patients into active groups (Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) score ≥ 6) and inactive groups (SLEDAI score < 6). Serum concentrations of CTHRC1, interferon alpha, interleukin (IL)-28A and IL-28B were determined using an enzyme-linked immunosorbent assay in a group of 40 patients with SLE. Results were compared with those from 23 healthy controls. RESULTS Serum CTHRC1 protein levels were higher in patients with SLE compared with healthy controls. Patients with active disease displayed higher CTHRC1 levels compared with those with inactive disease as well. There was a positive association between serum CTHRC1 levels and SLEDAI and erythrocyte sedimentation rate, and a negative correlation with complement 3 and 4. Moreover, serum CTHRC1 levels were higher in SLE patients with arthritis and anemia compared with patients without the above-mentioned manifestations. CONCLUSIONS These findings indicate CTHRC1 probably plays an important part in the pathogenesis of SLE, and is positively associated with disease activity, while it also likely refers to the development of arthritis and anemia in SLE. Therefore, CTHRC1 may provide a novel research target and shed new light on the pathogenesis and therapy of SLE.
Collapse
Affiliation(s)
- Q Wu
- Department of Rheumatology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, P.R. China
| | - Q Yang
- Department of Rheumatology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, P.R. China
| | - H Sun
- Department of Rheumatology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, P.R. China
| |
Collapse
|
7
|
Soloviova K, Puliaiev M, Puliaev R, Puliaeva I, Via CS. Both perforin and FasL are required for optimal CD8 T cell control of autoreactive B cells and autoantibody production in parent-into-F1 lupus mice. Clin Immunol 2018; 194:34-42. [PMID: 29940333 PMCID: PMC6089648 DOI: 10.1016/j.clim.2018.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 11/29/2022]
Abstract
To test the relative roles of perforin (pfp) vs. FasL in CTL control of autoreactive B cell expansion, we used the parent-into-F1 model of murine graft-vs.-host disease in which donor CD8 CTL prevent lupus like disease by eliminating activated autoreactive B cells. F1 mice receiving either pfp or FasL defective donor T cells exhibited an intermediate short-term phenotype. Pairing of purified normal CD4 T cells with either pfp or FasL defective CD8 T cell subsets resulted in impaired host B cell elimination and mild lupus like disease that was roughly equivalent in the two experimental groups. Thus, in addition to major roles in tumor and intracellular pathogen control, pfp mediated CD8 CTL killing plays a significant role in controlling autoreactive B cell expansion and lupus downregulation that is comparable to that mediated by FasL killing. Importantly, both pathways are required for optimal elimination of activated autoreactive B cells.
Collapse
Affiliation(s)
- Kateryna Soloviova
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20815, United States
| | - Maksym Puliaiev
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20815, United States
| | - Roman Puliaev
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20815, United States
| | - Irina Puliaeva
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20815, United States
| | - Charles S Via
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20815, United States.
| |
Collapse
|
8
|
Castañeda-Delgado JE, Bastián-Hernandez Y, Macias-Segura N, Santiago-Algarra D, Castillo-Ortiz JD, Alemán-Navarro AL, Martínez-Tejada P, Enciso-Moreno L, Garcia-De Lira Y, Olguín-Calderón D, Trouw LA, Ramos-Remus C, Enciso-Moreno JA. Type I Interferon Gene Response Is Increased in Early and Established Rheumatoid Arthritis and Correlates with Autoantibody Production. Front Immunol 2017; 8:285. [PMID: 28373872 PMCID: PMC5357778 DOI: 10.3389/fimmu.2017.00285] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 02/28/2017] [Indexed: 12/21/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is an inflammatory debilitating disease that affects the joints in the early and productive phases of an individual’s life. Several cytokines have been linked to the disease pathogenesis and are known to contribute to the inflammatory state characteristic of RA. The participation of type I interferon (IFN) in the pathogenesis of the disease has been already described as well as the identity of the genes that are regulated by this molecule, which are collectively known as the type I IFN signature. These genes have several functions associated with apoptosis, transcriptional regulation, protein degradation, Th2 cell induction, B cell proliferation, etc. This article evaluated the expression of several genes of the IFN signature in different stages of disease and their correlation with the levels of anticitrullinated protein antibodies (ACPA) anticarbamylated protein (Anti-CarP) antibodies. Methods Samples from individuals with early and established RA, high-risk individuals (ACPA+ and ACPA−), and healthy controls were recruited at “Unidad de Artritis y Rheumatismo” (Rheumatism and Arthritis Unit) in Guadalajara Jalisco Mexico. Determinations of ACPA were made with Eurodiagnostica ACPA plus kit. Anti-CarP determinations were made according to previously described protocols. RNA was isolated, and purity and integrity were determined according to RNA integrity number >6. Gene expression analysis was made by RT-qPCR using specific primers for mRNAs of the type I IFN signature. Relative gene expression was calculated according to Livak and Schmitgen. Results Significant differences in gene expression were identified when comparing the different groups for MXA and MXB (P < 0.05), also when comparing established RA and ACPA− in both IFIT 1 and G15. An increased expression of ISG15 was identified (P < 0.05), and a clear tendency toward increase was identified for HERC5. EPSTRI1, IFI6, and IFI35 were found to be elevated in the chronic/established RA and early RA (P < 0.05). Significant correlations were identified for the IFN signature genes with the levels of ACPA and anti-CarP (P < 0.05). Conclusion Our data confirm previous observations in the role of IFN signature and the pathogenesis of RA. Also, we provide evidence of an association between several genes of the IFN signature (that regulate Th2 cells and B cell proliferation) with the levels of anti-CarP antibodies and ACPA.
Collapse
Affiliation(s)
- Julio E Castañeda-Delgado
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS, Zacatecas, Mexico; National Council of Science and Technology, CONACYT, Catedras-CONACYT, Zacatecas, Mexico
| | - Yadira Bastián-Hernandez
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS, Zacatecas, Mexico; National Council of Science and Technology, CONACYT, Catedras-CONACYT, Zacatecas, Mexico
| | - Noe Macias-Segura
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS, Zacatecas, Mexico; Departamento de fisiología y farmacología, centro de ciencias básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Aguascalientes, Mexico
| | - David Santiago-Algarra
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| | - Jose D Castillo-Ortiz
- Unidad de Investigación en Enfermedades Crónico-Degenerativas , Guadalajara, Jalisco , México
| | - Ana L Alemán-Navarro
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| | - Pedro Martínez-Tejada
- General Hospital: "Emilio Varela Lujan", Mexican Institute of Social Security, IMSS , Zacatecas , Mexico
| | - Leonor Enciso-Moreno
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| | - Yolanda Garcia-De Lira
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| | - Diana Olguín-Calderón
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| | - Leendert A Trouw
- Department of Rheumatology, Leiden University Medical Center , Leiden , Netherlands
| | | | - Jose A Enciso-Moreno
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| |
Collapse
|
9
|
Doran AG, Wong K, Flint J, Adams DJ, Hunter KW, Keane TM. Deep genome sequencing and variation analysis of 13 inbred mouse strains defines candidate phenotypic alleles, private variation and homozygous truncating mutations. Genome Biol 2016; 17:167. [PMID: 27480531 PMCID: PMC4968449 DOI: 10.1186/s13059-016-1024-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 07/12/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The Mouse Genomes Project is an ongoing collaborative effort to sequence the genomes of the common laboratory mouse strains. In 2011, the initial analysis of sequence variation across 17 strains found 56.7 M unique single nucleotide polymorphisms (SNPs) and 8.8 M indels. We carry out deep sequencing of 13 additional inbred strains (BUB/BnJ, C57BL/10J, C57BR/cdJ, C58/J, DBA/1J, I/LnJ, KK/HiJ, MOLF/EiJ, NZB/B1NJ, NZW/LacJ, RF/J, SEA/GnJ and ST/bJ), cataloguing molecular variation within and across the strains. These strains include important models for immune response, leukaemia, age-related hearing loss and rheumatoid arthritis. We now have several examples of fully sequenced closely related strains that are divergent for several disease phenotypes. RESULTS Approximately 27.4 M unique SNPs and 5 M indels are identified across these strains compared to the C57BL/6 J reference genome (GRCm38). The amount of variation found in the inbred laboratory mouse genome has increased to 71 M SNPs and 12 M indels. We investigate the genetic basis of highly penetrant cancer susceptibility in RF/J finding private novel missense mutations in DNA damage repair and highly cancer associated genes. We use two highly related strains (DBA/1J and DBA/2J) to investigate the genetic basis of collagen-induced arthritis susceptibility. CONCLUSIONS This paper significantly expands the catalogue of fully sequenced laboratory mouse strains and now contains several examples of highly genetically similar strains with divergent phenotypes. We show how studying private missense mutations can lead to insights into the genetic mechanism for a highly penetrant phenotype.
Collapse
Affiliation(s)
- Anthony G Doran
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Kim Wong
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Jonathan Flint
- The Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - David J Adams
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Kent W Hunter
- Laboratory of Cancer Biology and Genetics, NCI, NIH, Bethesda, Maryland, USA.
| | - Thomas M Keane
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK.
| |
Collapse
|
10
|
Soloviova K, Puliaiev M, Haas M, Dalgard CL, Schaefer BC, Via CS. Intrinsic Differences in Donor CD4 T Cell IL-2 Production Influence Severity of Parent-into-F1 Murine Lupus by Skewing the Immune Response Either toward Help for B Cells and a Sustained Autoantibody Response or toward Help for CD8 T Cells and a Downregulatory Th1 Response. THE JOURNAL OF IMMUNOLOGY 2015; 195:2985-3000. [PMID: 26320249 DOI: 10.4049/jimmunol.1402782] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 07/28/2015] [Indexed: 01/06/2023]
Abstract
Using the parent-into-F1 model of induced lupus and (C57BL/6 × DBA2) F1 mice as hosts, we compared the inherent lupus-inducing properties of the two parental strain CD4 T cells. To control for donor CD4 recognition of alloantigen, we used H-2(d) identical DBA/2 and B10.D2 donor T cells. We demonstrate that these two normal, nonlupus-prone parental strains exhibit two different T cell activation pathways in vivo. B10.D2 CD4 T cells induce a strong Th1/CMI pathway that is characterized by IL-2/IFN-γ expression, help for CD8 CTLs, and skewing of dendritic cell (DC) subsets toward CD8a DCs, coupled with reduced CD4 T follicular helper cells and transient B cell help. In contrast, DBA/2 CD4 T cells exhibit a reciprocal, lupus-inducing pathway that is characterized by poor IL-2/IFN-γ expression, poor help for CD8 CTLs, and skewing of DC subsets toward plasmacytoid DCs, coupled with greater CD4 T follicular helper cells, prolonged B cell activation, autoantibody formation, and lupus-like renal disease. Additionally, two distinct in vivo splenic gene-expression signatures were induced. In vitro analysis of TCR signaling revealed defective DBA CD4 T cell induction of NF-κB, reduced degradation of IκBα, and increased expression of the NF-κB regulator A20. Thus, attenuated NF-κB signaling may lead to diminished IL-2 production by DBA CD4 T cells. These results indicate that intrinsic differences in donor CD4 IL-2 production and subsequent immune skewing could contribute to lupus susceptibility in humans. Therapeutic efforts to skew immune function away from excessive help for B cells and toward help for CTLs may be beneficial.
Collapse
Affiliation(s)
- Kateryna Soloviova
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Maksym Puliaiev
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Mark Haas
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Clifton L Dalgard
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814; and
| | - Brian C Schaefer
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Charles S Via
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814;
| |
Collapse
|
11
|
Zhang L, Bertucci AM, Ramsey-Goldman R, Harsha-Strong ER, Burt RK, Datta SK. Major pathogenic steps in human lupus can be effectively suppressed by nucleosomal histone peptide epitope-induced regulatory immunity. Clin Immunol 2013; 149:365-78. [PMID: 24211843 DOI: 10.1016/j.clim.2013.08.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/22/2013] [Accepted: 08/14/2013] [Indexed: 02/05/2023]
Abstract
Low-dose tolerance therapy with nucleosomal histone peptide epitopes blocks lupus disease in mouse models, but effect in humans is unknown. Herein, we found that CD4(+)CD25(high)FoxP3(+) or CD4(+)CD45RA(+)FoxP3(low) T-cells, and CD8(+)CD25(+)FoxP3(+) T-cells were all induced durably in PBMCs from inactive lupus patients and healthy subjects by the histone peptide/s themselves, but in active lupus, dexamethasone or hydroxychloroquine unmasked Treg-induction by the peptides. The peptide-induced Treg depended on TGFβ/ALK-5/pSmad 2/3 signaling, and they expressed TGF-β precursor LAP. Lupus patients' sera did not inhibit Treg induction. The peptide epitope-induced T cells markedly suppressed type I IFN related gene expression in lupus PBMC. Finally, the peptide epitopes suppressed pathogenic autoantibody production by PBMC from active lupus patients to baseline levels by additional mechanisms besides Treg induction, and as potently as anti-IL6 antibody. Thus, low-dose histone peptide epitopes block pathogenic autoimmune response in human lupus by multiple mechanisms to restore a stable immunoregulatory state.
Collapse
Affiliation(s)
- Li Zhang
- Divisions of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|
12
|
Singh RR, Yang JQ, Kim PJ, Halder RC. Germline deletion of β2 microglobulin or CD1d reduces anti-phospholipid antibody, but increases autoantibodies against non-phospholipid antigens in the NZB/W F1 model of lupus. Arthritis Res Ther 2013; 15:R47. [PMID: 23531237 PMCID: PMC3672782 DOI: 10.1186/ar4206] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 03/19/2013] [Indexed: 11/25/2022] Open
Abstract
Introduction β2-microglobulin (β2m) is required for the surface expression of MHC class I and class I-like proteins such as CD1d, Qa1 and neonatal Fc receptor (FcRn), all of which may impact the development of autoimmunity. Since CD1d is known to bind and present phospholipid antigens to T cells, we asked if the deficiency of β2m or CD1d will impact the development of anti-phospholipid antibodies as compared to other aspects of lupus autoimmunity. Methods We introgressed the β2m-null genotype onto the NZB and NZW backgrounds for 12 to 14 generations to generate genetically lupus-susceptible (NZB/NZW)F1 (BWF1) mice that are β2m-deficient (β2m°). Circulating immunoglobulins (Ig), rheumatoid factor (RF), anti-DNA and anti-cardiolipin (anti-CL) antibodies, and renal disease were analyzed in these and CD1d-deficient (CD1d°) BWF1 mice that we had previously generated. Results Whereas β2m° BWF1 mice had reduced serum IgG, they had increased mortality, nephritis, serum IgG anti-DNA antibody and RF as compared to heterozygous and wild-type littermates. These effects were recapitulated in CD1d° BWF1 mice, except that they also had increased serum IgG as compared to control littermates. Intriguingly, both β2m° and CD1d° mice had lower serum anti-CL antibody levels than in control littermates. Such CD1d dependence of anti-CL antibody production is not mediated by CD1d/glycolipid-reactive iNKT cells, as these cells reduced the production of RF and anti-DNA antibodies but had no effect on anti-CL antibodies. Conclusions We report a novel dichotomous role of β2m and CD1d, whereby these molecules differently regulate autoimmunity against phospholipid versus non-phospholipid autoantigens.
Collapse
|
13
|
Abstract
The characterization of functional CD8(+) inhibitory or regulatory T cells and their gene regulation remains a critical challenge in the field of tolerance and autoimmunity. Investigating the genes induced in regulatory cells and the regulatory networks and pathways that underlie mechanisms of immune resistance and prevent apoptosis in the CD8(+) T cell compartment are crucial to understanding tolerance mechanisms in systemic autoimmunity. Little is currently known about the genetic control that governs the ability of CD8(+) Ti or regulatory cells to suppress anti-DNA Ab production in B cells. Silencing genes with siRNA or shRNA and overexpression of genes with lentiviral cDNA transduction are established approaches to identifying and understanding the function of candidate genes in tolerance and immunity. Elucidation of interactions between genes and proteins, and their synergistic effects in establishing cell-cell cross talk, including receptor modulation/antagonism, are essential for delineating the roles of these cells. In this review, we will examine recent reports which describe the modulation of cells from lupus prone mice or lupus patients to confer anti-inflammatory and protective gene expression and novel associated phenotypes. We will highlight recent findings on the role of selected genes induced by peptide tolerance in CD8(+) Ti.
Collapse
|
14
|
Wen X, Yang JQ, Kim PJ, Singh RR. Homeostatic regulation of marginal zone B cells by invariant natural killer T cells. PLoS One 2011; 6:e26536. [PMID: 22046304 PMCID: PMC3202546 DOI: 10.1371/journal.pone.0026536] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 09/28/2011] [Indexed: 11/18/2022] Open
Abstract
Marginal zone B cells (MZB) mount a rapid antibody response, potently activate naïve T cells, and are enriched in autoreactive B cells. MZBs express high levels of CD1d, the restriction element for invariant natural killer T cells (iNKT). Here, we examined the effect of iNKT cells on MZB cell activation and numbers in vitro and in vivo in normal and autoimmune mice. Results show that iNKT cells activate MZBs, but restrict their numbers in vitro and in vivo in normal BALB/c and C57/BL6 mice. iNKT cells do so by increasing the activation-induced cell death and curtailing proliferation of MZB cells, whereas they promote the proliferation of follicular B cells. Sorted iNKT cells can directly execute this function, without help from other immune cells. Such MZB regulation by iNKTs is mediated, at least in part, via CD1d on B cells in a contact-dependent manner, whereas iNKT-induced proliferation of follicular B cells occurs in a contact- and CD1d-independent manner. Finally, we show that iNKT cells reduce 'autoreactive' MZB cells in an anti-DNA transgenic model, and limit MZB cell numbers in autoimmune-prone (NZB×NZW)F1 and non-obese diabetic mice, suggesting a potentially new mechanism whereby iNKT cells might regulate pathologic autoimmunity. Differential regulation of follicular B cells versus potentially autoreactive MZBs by iNKT cells has important implications for autoimmune diseases as well as for conditions that require a rapid innate B cell response.
Collapse
Affiliation(s)
- Xiangshu Wen
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Jun-Qi Yang
- University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
| | - Peter J. Kim
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Ram Raj Singh
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
15
|
Sawla P, Hossain A, Hahn BH, Singh RP. Regulatory T cells in systemic lupus erythematosus (SLE); role of peptide tolerance. Autoimmun Rev 2011; 11:611-4. [PMID: 22001419 DOI: 10.1016/j.autrev.2011.09.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Accepted: 09/27/2011] [Indexed: 12/23/2022]
Abstract
Regulatory T cells play an important role in the maintenance and regulation of immune tolerance and in the prevention of autoimmunity. Recent studies have demonstrated a deficiency in number and function of regulatory T cells in lupus and other autoimmune diseases. This may contribute to immune dysregulations and a defect in self-tolerance mechanisms. How to balance and "reset" the immune response from harmful pro-inflammatory to beneficial anti-inflammatory is the current strategy of the research. In this regard, several studies have been performed with various peptides, drugs, steroids and epigenetic agents to induce or modify regulatory cells and some measure of success has been achieved in the animal model of SLE and with lupus patient cells. Challenges ahead include the heterogeneous nature, phenotype and function of regulatory cells and the difficulties in manipulation of regulatory function in healthy versus diseased states. In this review, we will provide some recent findings indicating challenges and potential benefits of targeting of regulatory T cells in lupus.
Collapse
Affiliation(s)
- Priya Sawla
- Division of Rheumatology, Dept. of Medicine, David Geffen School of Medicine, University of California at Los Angeles, CA 90095-1670, USA
| | | | | | | |
Collapse
|
16
|
Kang HK, Chiang MY, Liu M, Ecklund D, Datta SK. The histone peptide H4 71-94 alone is more effective than a cocktail of peptide epitopes in controlling lupus: immunoregulatory mechanisms. J Clin Immunol 2011; 31:379-94. [PMID: 21287397 PMCID: PMC3125430 DOI: 10.1007/s10875-010-9504-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 12/27/2010] [Indexed: 12/01/2022]
Abstract
Tolerance therapy with nucleosomal histone peptides H4(71-94), H4(16-39), or H1'(22-42) controls disease in lupus-prone SNF1 mice. It would be clinically important to determine whether a cocktail of the above epitopes would be superior. Herein, we found that compared with cocktail peptides, H4(71-94) monotherapy more effectively delayed nephritis onset, prolonged lifespan, diminished immunoglobulin G autoantibody levels, reduced autoantigen-specific Th1 and Th17 responses and frequency of T(FH) cells in spleen and the helper ability of autoimmune T cells to B cells, by inducing potent CD8 Treg cells. H4(71-94) therapy was superior in "tolerance spreading," suppressing responses to other autoepitopes, nucleosomes, and ribonucleoprotein. We also developed an in vitro assay for therapeutic peptides (potentially in humans), which showed that H4(71-94), without exogenous transforming growth factor (TGF)-β, was efficient in inducing stable CD4(+)CD25(+)Foxp3(+) T cells by decreasing interleukin 6 and increasing TGF-β production by dendritic cells that induced ALK5-dependent Smad-3 phosphorylation (TGF-β signal) in target autoimmune CD4(+) T cells.
Collapse
Affiliation(s)
- Hee-Kap Kang
- Division of Rheumatology, Departments of Medicine, Northwestern University Feinberg School of Medicine, 240 East Huron St. McGaw # M300, Chicago, IL 60611, USA
| | - Ming-Yi Chiang
- Division of Rheumatology, Departments of Medicine, Northwestern University Feinberg School of Medicine, 240 East Huron St. McGaw # M300, Chicago, IL 60611, USA
| | - Michael Liu
- Division of Rheumatology, Departments of Medicine, Northwestern University Feinberg School of Medicine, 240 East Huron St. McGaw # M300, Chicago, IL 60611, USA
| | - Diane Ecklund
- Division of Rheumatology, Departments of Medicine, Northwestern University Feinberg School of Medicine, 240 East Huron St. McGaw # M300, Chicago, IL 60611, USA
| | - Syamal K. Datta
- Division of Rheumatology, Departments of Medicine, Northwestern University Feinberg School of Medicine, 240 East Huron St. McGaw # M300, Chicago, IL 60611, USA
| |
Collapse
|
17
|
Resistance to exogenous TGF-β effects in patients with systemic lupus erythematosus. J Clin Immunol 2011; 31:574-83. [PMID: 21503670 DOI: 10.1007/s10875-011-9531-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 04/03/2011] [Indexed: 12/23/2022]
Abstract
BACKGROUND The mechanisms underlying the loss of self-tolerance in systemic lupus erythematosus (SLE) are incompletely deciphered. TGF-β plays a key role in self-tolerance demonstrated by the onset of a fatal autoimmune syndrome associated with lupus autoantibodies in mice lacking a functional TGF-β receptor. The present work aims to define whether resistance to TGF-β might contribute to the pathogenesis of SLE. METHODS Twenty-two patients with active SLE, 16 with other connective tissue diseases, and 10 healthy controls were prospectively included in this study. The effects of exogenous TGF-β1 on IL-2-dependent T-cell proliferation, IFN-γ secretion, and target gene transcription were analyzed on peripheral blood mononuclear cells. RESULTS Our results showed that 75% of patients with SLE or other connective tissue diseases were totally or partially resistant to the effects of TGF-β1. The responses to the anti-proliferative and transcriptional effects of TGF-β were, however, discordant in a high proportion of our patients. Hence, we distinguish three distinct profiles of resistance to TGF-β1 and suggest that patients may exhibit different defects affecting distinct points of TGF-β1 signaling pathways. CONCLUSION Our data demonstrate the presence of an impaired response of peripheral cells to TGF-β1 in patients with active SLE that may participate to the pathogenesis of the disease. Further studies will be necessary to delineate the mechanisms underlying the lymphocyte resistance to TGF-β1 in SLE.
Collapse
|
18
|
Liu SD, Lee S, La Cava A, Motran CC, Hahn BH, Miceli MC. Galectin-1-induced down-regulation of T lymphocyte activation protects (NZB x NZW) F1 mice from lupus-like disease. Lupus 2011; 20:473-84. [PMID: 21335401 DOI: 10.1177/0961203310388444] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by a hyperactive immune system, including activation of autoreactive T and B cells. These studies demonstrate that administration of recombinant galectin-1, a β-galactose binding protein, to SLE-prone (NZB × NZW) F1 mice reduced lymphocyte activation, inhibited serum anti-double-stranded DNA(dsDNA) IgG antibody production, decreased the incidence of proteinuria, and increased survival rate. In addition, recombinant galectin-1'-treated mice had a higher frequency of Foxp3 expression, which suggested an increase in the percentage of peripheral regulatory T cells. Consistent with the finding that there were fewer activated T lymphocytes, ex vivo T cells from mice treated with recombinant galectin-1 exhibited less proliferation in response to TCR stimulation. Furthermore, these cells were less efficient at lipid raft clustering in response to TCR/CD28 engagement, consistent with published reports that galectin-1 can reorganize the synaptic contact to interfere with TCR signaling and activation to prevent T cell activation. Aged galectin-1-deficient mice had higher serum levels of antibodies against dsDNA, elucidating a role for endogenous galectin-1 in decreasing susceptibility to autoimmunity. Together, the findings highlight galectin-1 as a novel potential therapeutic immune modulator for treatment of lupus-like disease.
Collapse
Affiliation(s)
- S D Liu
- University of California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
19
|
Dinesh R, Hahn BH, La Cava A, Singh RP. Interferon-inducible gene 202b controls CD8(+) T cell-mediated suppression in anti-DNA Ig peptide-treated (NZB × NZW) F1 lupus mice. Genes Immun 2011; 12:360-9. [PMID: 21326316 PMCID: PMC3149980 DOI: 10.1038/gene.2011.4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Administration of an artificial peptide (pConsensus) based on anti-DNA IgG sequences that contain major histocompatibility complex class I and class II T-cell determinants, induces immune tolerance in NZB/NZW F1 female (BWF1) mice. To understand the molecular basis of CD8+ Ti-mediated suppression, we previously performed microarray analysis to identify genes that were differentially expressed following tolerance induction with pCons. CD8+ T cells from mice tolerized with pCons showed more than two-fold increase in Ifi202b mRNA, an interferon inducible gene, versus cells from untolerized mice. Ifi202b expression increased through weeks 1–4 after tolerization and then decreased, reapproaching baseline levels at 6 weeks. In vitro polyclonal activation of tolerized CD8+ T cells significantly increased Ifi202b mRNA expression. Importantly, silencing of Ifi202b abrogated the suppressive capacity of CD8+ Ti cells. This was associated with decreased expression of Foxp3, and decreased gene and protein expression of transforming growth factor (TGF)β and interleukin-2 (IL-2), but not of interferon (IFN)-γ, IL-10, or IL-17. Silencing of another IFN-induced gene upregulated in tolerized CD8+ T cells, IFNAR1, had no effect on the ability of CD8+ T cells to suppress autoantibody production. Our findings indicate a potential role for Ifi202b in the suppressive capacity of peptide-induced regulatory CD8+ Ti cells through effects on the expression of Foxp3 and the synthesis of TGFβ.
Collapse
Affiliation(s)
- R Dinesh
- Division of Rheumatology, Department of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
20
|
Divekar AA, Dubey S, Gangalum PR, Singh RR. Dicer insufficiency and microRNA-155 overexpression in lupus regulatory T cells: an apparent paradox in the setting of an inflammatory milieu. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:924-30. [PMID: 21149603 PMCID: PMC3038632 DOI: 10.4049/jimmunol.1002218] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Systemic lupus erythematosus is a chronic autoimmune disease characterized by loss of tolerance to self-Ags and activation of autoreactive T cells. Regulatory T (Treg) cells play a critical role in controlling the activation of autoreactive T cells. In this study, we investigated mechanisms of potential Treg cell defects in systemic lupus erythematosus using MRL-Fas(lpr/lpr) (MRL/lpr) and MRL-Fas(+/+) mouse models. We found a significant increase in CD4(+)CD25(+)Foxp3(+) Treg cells, albeit with an altered phenotype (CD62L(-)CD69(+)) and with a reduced suppressive capacity, in the lymphoid organs of MRL strains compared with non-autoimmune C3H/HeOuj mice. A search for mechanisms underlying the altered Treg cell phenotype in MRL/lpr mice led us to find a profound reduction in Dicer expression and an altered microRNA (miRNA, miR) profile in MRL/lpr Treg cells. Despite having a reduced level of Dicer, MRL/lpr Treg cells exhibited a significant overexpression of several miRNAs, including let-7a, let-7f, miR-16, miR-23a, miR-23b, miR-27a, and miR-155. Using computational approaches, we identified one of the upregulated miRNAs, miR-155, that can target CD62L and may thus confer the altered Treg cell phenotype in MRL/lpr mice. In fact, the induced overexpression of miR-155 in otherwise normal (C3H/HeOuj) Treg cells reduced their CD62L expression, which mimics the altered Treg cell phenotype in MRL/lpr mice. These data suggest a role of Dicer and miR-155 in regulating Treg cell phenotype. Furthermore, simultaneous appearance of Dicer insufficiency and miR-155 overexpression in diseased mice suggests a Dicer-independent alternative mechanism of miRNA regulation under inflammatory conditions.
Collapse
Affiliation(s)
- Anagha A. Divekar
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Departments of Medicine and of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095
| | - Shweta Dubey
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Departments of Medicine and of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095
| | - Pallavi R. Gangalum
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Departments of Medicine and of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095
| | - Ram Raj Singh
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Departments of Medicine and of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
21
|
Implications of the parent-into-F1 model for human lupus pathogenesis: roles for cytotoxic T lymphocytes and viral pathogens. Curr Opin Rheumatol 2010; 22:493-8. [PMID: 20485174 DOI: 10.1097/bor.0b013e32833b0174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW The central role of CD4 T cells in lupus pathogenesis is well recognized; however, the mechanism by which CD4 T cells lose tolerance and promote humoral autoimmunity remains unclear. This review examines mechanisms elucidated in the parent-into-F1 model of lupus and their possible parallels in human lupus pathogenesis. RECENT FINDINGS In the parent-into-F1 model, lupus results from the transfer of normal, foreign reactive CD4 T cells targeted to intrinsically normal F1 B cells. Transfer of normal CD8 T cells prevents lupus, whereas transfer of CD8 T cells with killing defects does not but is correctable with additional in-vivo enhancement of CD8 cytotoxic T lymphocyte (CTL) function. The parent-into-F1 model has two major similarities to Epstein-Barr virus infection: CD4 T-cell-driven polyclonal B-cell hyperactivity and a critical dependence on CD8 CTL for elimination of activated B cells. These similarities are discussed in relation to human lupus pathogenesis. SUMMARY Work in the parent-into-F1 model supports the idea that lupus may result from defective CD8 T-cell function and that therapeutic enhancement of CD8 effectors with selective targeting to autoreactive B cells may be beneficial. Despite strong evidence linking Epstein-Barr virus infection with human lupus, the exact nature of this link requires further study.
Collapse
|
22
|
Dinesh RK, Skaggs BJ, Cava AL, Hahn BH, Singh RP. CD8+ Tregs in lupus, autoimmunity, and beyond. Autoimmun Rev 2010; 9:560-8. [PMID: 20385256 PMCID: PMC2879466 DOI: 10.1016/j.autrev.2010.03.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Accepted: 03/30/2010] [Indexed: 12/12/2022]
Abstract
While CD4(+)CD25(high) regulatory T cells (Tregs) have garnered much attention for their role in the maintenance of immune homeostasis, recent findings have shown that subsets of CD8(+) T cells (CD8(+) Tregs) display immunoregulatory functions as well. Both CD4(+) Tregs and CD8(+) Tregs appear impaired in number and/or function in several autoimmune diseases and in experimental animal models of autoimmunity, suggesting the possibility of immunotherapeutic targeting of these cells for improved management of autoimmune conditions. Our group has developed a strategy to induce CD8(+) Tregs in autoimmune mice through the use of a tolerogenic self-peptide, and new information has been gained on the phenotype, function and role of induced CD8(+) Tregs in autoimmunity. Here we present an overview of the role and mechanisms of action of CD8(+) Tregs in autoimmunity, with a special focus on lupus. We also discuss the potential role of CD8(+) Tregs in other diseases, including chronic infection and cancer.
Collapse
Affiliation(s)
- Ravi K Dinesh
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1670
| | - Brian J Skaggs
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1670
| | - Antonio La Cava
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1670
| | - Bevra H. Hahn
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1670
| | - Ram Pyare Singh
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1670
| |
Collapse
|
23
|
Singh RP, Dinesh R, Elashoff D, de Vos S, Rooney RJ, Patel D, La Cava A, Hahn BH. Distinct gene signature revealed in white blood cells, CD4(+) and CD8(+) T cells in (NZBx NZW) F1 lupus mice after tolerization with anti-DNA Ig peptide. Genes Immun 2010; 11:294-309. [PMID: 20200542 PMCID: PMC10725082 DOI: 10.1038/gene.2010.6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Revised: 11/27/2009] [Accepted: 01/06/2010] [Indexed: 11/08/2022]
Abstract
Tolerizing mice polygenically predisposed to lupus-like disease (NZB/NZW F1 females) with a peptide mimicking anti-DNA IgG sequences containing MHC class I and class II T cell determinants (pConsensus, pCons) results in protection from full-blown disease attributable in part to the induction of CD4(+)CD25(+)Foxp3+ and CD8(+)Foxp3+ regulatory T cells. We compared 45 000 murine genes in total white blood cells (WBC), CD4(+) T cells, and CD8(+) T cells from splenocytes of (NZBxNZW) F1 lupus-prone mice tolerized with pCons vs untreated naïve mice and found two-fold or greater differential expression for 448 WBC, 174 CD4, and 60 CD8 genes. We identified differentially expressed genes that played roles in the immune response and apoptosis. Using real-time PCR, we validated differential expression of selected genes (IFI202B, Bcl2, Foxp3, Trp-53, CCR7 and IFNar1) in the CD8(+)T cell microarray and determined expression of selected highly upregulated genes in different immune cell subsets. We also determined Smads expression in different immune cell subsets, including CD4(+) T cells and CD8(+) T cells, to detect the effects of TGF-beta, known to be the major cytokine that accounts for the suppressive capacity of CD8(+) Treg in this system. Silencing of anti-apoptotic gene Bcl2 or interferon genes (IFI202b and IFNar1 in combination) in CD8(+) T cells from tolerized mice did not affect the expression of the other selected genes. However, silencing of Foxp3 reduced expression of Foxp3, Ifi202b and PD1-all of which are involved in the suppressive capacity of CD8(+) Treg in this model.
Collapse
Affiliation(s)
- R P Singh
- Division of Rheumatology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Hirabayashi Y, Oka Y, Ikeda T, Fujii H, Ishii T, Sasaki T, Harigae H. The endoplasmic reticulum stress-inducible protein, Herp, is a potential triggering antigen for anti-DNA response. THE JOURNAL OF IMMUNOLOGY 2010; 184:3276-83. [PMID: 20147634 DOI: 10.4049/jimmunol.0900670] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anti-dsDNA Abs are highly specific indicators of systemic lupus erythematosus (SLE) and play a pathogenic role in lupus nephritis. Human anti-dsDNA Abs are most likely generated by an Ag-driven mechanism. However, the Ag responsible for triggering anti-dsDNA Ab production has not been identified. To search for proteins that are cross-reactive with anti-dsDNA Abs, we screened a cDNA library from a patient with SLE with single-chain Fv of O-81 human anti-ss/dsDNA mAb by using a two-hybrid system. Homocysteine-induced ER protein (Herp), an endoplasmic reticulum (ER) stress-inducible ER membrane protein, was identified and shown to bind to original O-81 Ab and human lupus anti-dsDNA Abs. Some IgG purified from patients with active SLE by Herp-immobilized affinity chromatography bound to dsDNA. BALB/c mice immunized with Herp showed IgG anti-dsDNA Abs, IgG anti-nucleosome Abs, and glomerular IgG deposition. Herp reactivity was strongly positive in a proportion of PBLs from patients with active SLE, but undetectable in those from healthy controls. Moreover, activation of caspases was observed in the Herp-positive cells, implying that ER stress-induced apoptosis likely occurs in patients with active SLE. Herp is exposed on blebs of ER stress-induced apoptotic cells, suggesting that Herp can be recognized by immune cells. These results indicate that Herp mimics structural determinants of DNA immunologically and can be immunogenic in vivo. Thus, Herp represents a candidate autoantigen for anti-DNA Abs. This study may help explain how common environmental factors induce the production of anti-DNA Abs and contribute the development of SLE.
Collapse
Affiliation(s)
- Yasuhiko Hirabayashi
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | | | | | | | | | | | | |
Collapse
|
25
|
Guth A, Detanico T, Smith D, Tung KS, Bonorino C, Wysocki LJ. Spontaneous autoimmunity in mice that carry an IghV partial transgene: a required arginine in VHCDR3. Lupus 2009; 18:299-308. [PMID: 19276297 PMCID: PMC2892212 DOI: 10.1177/0961203308097480] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We describe a unique spontaneous mouse model of autoimmunity, which occurs on a non-autoimmune-prone SWR genetic background. In this model, SWR mice carry an IghV partial transgene (pTg) encoding only the heavy chain variable domain of an antibody directed against chromatin. Autoimmune disease in pTg mice was manifested by some of the features of systemic lupus erythematosus (SLE), including the presence of serum anti-nuclear antibodies, splenomegaly, skin lesions and a moderate degree of kidney pathology, in various combinations among individuals. Autoimmunity was observed in three independent transgenic lines, but not in three control lines carrying a nearly identical pTg, in which a VHCDR3 codon for Arg was replaced by one for Ser to ablate chromatin reactivity. Various features of disease were often but not always accompanied by anti-chromatin antibodies. Unexpectedly, the anti-chromatin antibodies detected in seropositive animals were not encoded by the pTg. These observations strongly implicate a role for the transgene product in disease initiation but not necessarily for end-state pathology, and they raise the possibility that autoreactive B cells may play a previously unappreciated role in initiating the development of systemic autoimmunity.
Collapse
Affiliation(s)
- Amanda Guth
- Integrated Department of Immunology, National Jewish Medical and Research Center, and University of Colorado School of Medicine, Denver, CO 80206, USA
| | - Thiago Detanico
- Integrated Department of Immunology, National Jewish Medical and Research Center, and University of Colorado School of Medicine, Denver, CO 80206, USA
- Faculdade de Biociências and Instituto de Pesquisasx Biomédicas, PUCRS, 90010, Porto Alegre, RS, Brazil
| | - Diana Smith
- Integrated Department of Immunology, National Jewish Medical and Research Center, and University of Colorado School of Medicine, Denver, CO 80206, USA
| | - Kenneth S.K. Tung
- Departments of Microbiology and Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Cristina Bonorino
- Faculdade de Biociências and Instituto de Pesquisasx Biomédicas, PUCRS, 90010, Porto Alegre, RS, Brazil
| | - Lawrence J. Wysocki
- Integrated Department of Immunology, National Jewish Medical and Research Center, and University of Colorado School of Medicine, Denver, CO 80206, USA
| |
Collapse
|
26
|
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the production of autoantibodies that can form immune complexes and deposit in tissues, causing inflammation and organ damage. There is evidence that interferons and some interleukins can have an active role in the pathogenesis of SLE and can contribute significantly to the immune imbalance in the disease, whereas the role of some cytokines (such as TNF) is still debated. This review discusses the activity of several cytokines in SLE, their effects on the immune cells in relation to the disease pathogenesis, and the promise and limitations of cytokine-based therapies in clinical trials for lupus patients.
Collapse
Affiliation(s)
- Elaine V. Lourenço
- Division of Rheumatology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095-1670, USA
| | - Antonio La Cava
- Division of Rheumatology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095-1670, USA
| |
Collapse
|
27
|
Abstract
Recent evidence supports the idea that following a break in tolerance, CD8 cytotoxic T lymphocytes (CTL) may be an important but unrecognized mechanism for limiting expansion of autoreactive B cells. Failure of this mechanism could allow persistence of CD4 T cell driven polyclonal B cell activation resulting in clinical lupus. Although CD8 CTL failure may occur early in disease, work in mice supports the concept that therapeutic CTL enhancement may be both practical and beneficial in lupus. Devising such therapy for humans will first require an understanding of the in vivo mechanisms critical in CTL expansion and down regulation, particularly in the lupus setting which may differ from CTL generation in other clinical settings (e.g. tumors, infections).
Collapse
Affiliation(s)
- I. Puliaeva
- Pathology Department, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - R. Puliaev
- Pathology Department, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - C.S. Via
- Pathology Department, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| |
Collapse
|
28
|
Bommireddy R, Babcock GF, Singh RR, Doetschman T. TGFbeta1 deficiency does not affect the generation and maintenance of CD4+CD25+FOXP3+ putative Treg cells, but causes their numerical inadequacy and loss of regulatory function. Clin Immunol 2008; 127:206-13. [PMID: 18308639 PMCID: PMC2387055 DOI: 10.1016/j.clim.2007.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Revised: 11/27/2007] [Accepted: 12/18/2007] [Indexed: 10/22/2022]
Abstract
TGFbeta1 is considered to be required for peripheral maintenance of CD4(+)CD25(+)FOXP3(+) T(reg) cells. However, we demonstrate no reduction in the percentage of such T cells in the spleens and thymi of Tgfb1(-/-) mice. Although putative T(reg) cells, characterized as CD4(+)CD25(+)FOXP3(+)CD62L(+) T cells, are increased in Tgfb1(-/-) mice, they may be inadequate to control activated T cells since the ratio of activated T cells:putative T(reg) cells is several-fold higher in Tgfb1(-/-) mice than in control mice. We further show that whereas Tgfb1(-/-) mice that express a chicken OVA-specific TCR transgene (DO11.10) have an increase in putative T(reg) cells, there are no detectable CD4(+)CD25(+) T cells in the spleens of DO11.10 Rag1(-/-) mice suggesting that T(reg)-cell generation is self-antigen dependent regardless of whether they express Tgfb1. Finally, we demonstrate that Tgfb1(-/-) T cells remain responsive to the suppressive effect of TGFbeta1 in vitro. These data suggest that TGFbeta1 is required for the regulatory function of T(reg) cells to prevent activation of T cells and autoimmunity.
Collapse
Affiliation(s)
- Ramireddy Bommireddy
- BIO5 Institute, University of Arizona, PO Box 245217, Tucson, AZ 85724-5217, USA
- Department of Immunobiology, University of Arizona, PO Box 245217, Tucson, AZ 85724-5217, USA
| | - George F. Babcock
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Shriners Hospital for Children, Cincinnati, OH 45229, USA
| | - Ram R. Singh
- Department of Medicine/Rheumatology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Thomas Doetschman
- BIO5 Institute, University of Arizona, PO Box 245217, Tucson, AZ 85724-5217, USA
- Department of Cell Biology and Anatomy, University of Arizona, PO Box 245217, Tucson, AZ 85724-5217, USA
- Cancer Center, University of Arizona, PO Box 245217, Tucson, AZ 85724-5217, USA
| |
Collapse
|
29
|
Singh RP, La Cava A, Hahn BH. pConsensus peptide induces tolerogenic CD8+ T cells in lupus-prone (NZB x NZW)F1 mice by differentially regulating Foxp3 and PD1 molecules. THE JOURNAL OF IMMUNOLOGY 2008; 180:2069-80. [PMID: 18250412 DOI: 10.4049/jimmunol.180.4.2069] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Systemic lupus erythematosus is an autoimmune disease caused primarily by autoantibodies (including IgG anti-DNA) and immune complexes that cause tissue damage. After tolerization with an artificial peptide (pConsensus, pCons) based on murine anti-DNA IgG sequences containing MHC class I and class II T cell determinants, lupus-prone (NZB x NZW)F(1) female (BWF(1)) mice develop regulatory CD4+CD25+ T cells and inhibitory CD8+ T cells, both of which suppress anti-DNA Ig production and immune glomerulonephritis. In the present work, we show that splenocytes from BWF(1) mice treated with pCons had significant expansion of primarily CD8+ T cells. CD4+ T cells and B cells were each directly suppressed by CD8+ T cells from tolerized mice in a contact-independent manner. Both pCons-induced CD8+CD28+ and CD8+CD28- T cells suppressed production of anti-DNA in vitro. Silencing with small interfering RNA of Foxp3 abrogated the suppression mediated by both CD8+ T cell subsets. Additionally, CD8+ T cells from tolerized mice were weakly cytotoxic against syngeneic B cells from old anti-DNA-producing mice, but not from young mice. Importantly, pCons treatment had dual effects on CD8+ suppressor T cells from tolerized mice, increasing the intracellular expression of Foxp3 while decreasing the surface expression of PD1 molecules. Blocking PD1/PDL1 interactions in the CD8+ T cells from tolerized mice reduced their expression of Foxp3 and their ability to suppress CD4+CD25- proliferation. In contrast, blocking PD1/PDL1 in naive T cells increased Foxp3 expression. Our data suggest that tolerization with pCons activates different subsets of inhibitory/cytotoxic CD8+ T cells whose targets are both CD4+CD25- effector T cells and B cells.
Collapse
Affiliation(s)
- Ram Pyare Singh
- Division of Rheumatology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, 1000 Veteran Avenue, Los Angeles, CA 90095, USA.
| | | | | |
Collapse
|
30
|
Zhang L, Bertucci AM, Smith KA, Xu L, Datta SK. Hyperexpression of cyclooxygenase 2 in the lupus immune system and effect of cyclooxygenase 2 inhibitor diet therapy in a murine model of systemic lupus erythematosus. ACTA ACUST UNITED AC 2008; 56:4132-41. [PMID: 18050205 DOI: 10.1002/art.23054] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To investigate the role of cyclooxygenase 2 (COX-2) in the functioning of different cell types involved in the lupus autoimmune response, and to examine the therapeutic effect of COX-2 inhibitors in mice prone to spontaneously develop systemic lupus erythematosus (SLE). METHODS Lupus-prone (SWR x NZB)F(1) mice were fed with a diet containing different doses of the COX-2-specific inhibitor celecoxib or the nonspecific inhibitor aspirin, or a combination of both, and the effects of the therapy on autoantibody production, development of lupus nephritis, and mortality were determined. Expression of COX-2 by different cells of the lupus immune system and the effect of COX-2 inhibitors on the function of these cells in vitro and in vivo were assessed. RESULTS The immune cells of mice with SLE spontaneously hyperexpressed COX-2, and COX-2 inhibitors could cause cell apoptosis. Treatment with COX-2 inhibitors resulted in decreased autoantibody production and inhibition of the T cell response to the major lupus autoantigen, nucleosome, and its presentation by antigen-presenting cells. Surprisingly, a significant increase in survival occurred only in mice receiving intermittent therapy with the lowest dose of celecoxib (500 parts per million), approximating <100 mg of celecoxib/day in humans. A continuous diet, but not intermittent feeding, with the combination of celecoxib and aspirin delayed development of nephritis temporarily, but failed to prolong survival. Indeed, treatment with aspirin alone increased mortality. CONCLUSION The contributions of the major players in the pathogenic autoimmune response, namely, T cells, B cells, dendritic cells, and macrophages that are abnormally hyperactive in lupus, depend on the increased expression and activity of COX-2, similar to inflammatory cells in target organs. Intermittent pulse therapy with low doses of select COX-2 inhibitors would be of value in the treatment of lupus.
Collapse
Affiliation(s)
- Li Zhang
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, 240 East Huron Street, Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
31
|
Saxena V, Lienesch DW, Zhou M, Bommireddy R, Azhar M, Doetschman T, Singh RR. Dual roles of immunoregulatory cytokine TGF-beta in the pathogenesis of autoimmunity-mediated organ damage. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:1903-12. [PMID: 18209088 PMCID: PMC2291535 DOI: 10.4049/jimmunol.180.3.1903] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ample evidence suggests a role of TGF-beta in preventing autoimmunity. Multiorgan inflammatory disease, spontaneous activation of self-reactive T cells, and autoantibody production are hallmarks of autoimmune diseases, such as lupus. These features are reminiscent of the immunopathology manifest in TGF-beta1-deficient mice. In this study, we show that lupus-prone (New Zealand Black and White)F(1) mice have reduced expression of TGF-beta1 in lymphoid tissues, and TGF-beta1 or TGF-beta1-producing T cells suppress autoantibody production. In contrast, the expression of TGF-beta1 protein and mRNA and TGF-beta signaling proteins (TGF-beta receptor type II and phosphorylated SMAD3) increases in the target organs, i.e., kidneys, of these mice as they age and develop progressive organ damage. In fact, the levels of TGF-beta1 in kidney tissue and urine correlate with the extent of chronic lesions that represent local tissue fibrosis. In vivo TGF-beta blockade by treatment of these mice with an anti-TGF-beta Ab selectively inhibits chronic fibrotic lesions without affecting autoantibody production and the inflammatory component of tissue injury. Thus, TGF-beta plays a dual, seemingly paradoxical, role in the development of organ damage in multiorgan autoimmune diseases. According to our working model, reduced TGF-beta in immune cells predisposes to immune dysregulation and autoantibody production, which causes tissue inflammation that triggers the production of anti-inflammatory cytokines such as TGF-beta in target organs to counter inflammation. Enhanced TGF-beta in target organs, in turn, can lead to dysregulated tissue repair, progressive fibrogenesis, and eventual end-organ damage.
Collapse
Affiliation(s)
- Vijay Saxena
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Douglas W. Lienesch
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Min Zhou
- Autoimmunity and Tolerance Laboratory, Department of Medicine, University of California, Los Angeles, CA 90095
| | - Ramireddy Bommireddy
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Mohamad Azhar
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Thomas Doetschman
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Ram Raj Singh
- Autoimmunity and Tolerance Laboratory, Department of Medicine, University of California, Los Angeles, CA 90095
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| |
Collapse
|
32
|
Kang HK, Liu M, Datta SK. Low-dose peptide tolerance therapy of lupus generates plasmacytoid dendritic cells that cause expansion of autoantigen-specific regulatory T cells and contraction of inflammatory Th17 cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:7849-58. [PMID: 17548623 DOI: 10.4049/jimmunol.178.12.7849] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Subnanomolar doses of an unaltered, naturally occurring nucleosomal histone peptide epitope, H4(71-94), when injected s.c. into lupus-prone mice, markedly prolong lifespan by generating CD4+25+ and CD8+ regulatory T cells (Treg) producing TGF-beta. The induced Treg cells suppress nuclear autoantigen-specific Th and B cells and block renal inflammation. Splenic dendritic cells (DC) captured the s.c.-injected H4(71-94) peptide rapidly and expressed a tolerogenic phenotype. The DC of the tolerized animal, especially plasmacytoid DC, produced increased amounts of TGF-beta, but diminished IL-6 on stimulation via the TLR-9 pathway by nucleosome autoantigen and other ligands; and those plasmacytoid DC blocked lupus autoimmune disease by simultaneously inducing autoantigen-specific Treg and suppressing inflammatory Th17 cells that infiltrated the kidneys of untreated lupus mice. Low-dose tolerance with H4(71-94) was effective even though the lupus immune system is spontaneously preprimed to react to the autoepitope. Thus, H4(71-94) peptide tolerance therapy that preferentially targets pathogenic autoimmune cells could spare lupus patients from chronically receiving toxic agents or global immunosuppressants and maintain remission by restoring autoantigen-specific Treg cells.
Collapse
Affiliation(s)
- Hee-Kap Kang
- Division of Rheumatology, Departments of Medicine and Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 240 East Huron Street, Chicago, IL 60611, USA
| | | | | |
Collapse
|
33
|
Yang JQ, Wen X, Liu H, Folayan G, Dong X, Zhou M, Van Kaer L, Singh RR. Examining the role of CD1d and natural killer T cells in the development of nephritis in a genetically susceptible lupus model. ARTHRITIS AND RHEUMATISM 2007; 56:1219-33. [PMID: 17393451 PMCID: PMC2291538 DOI: 10.1002/art.22490] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE CD1d-reactive invariant natural killer T (iNKT) cells secrete multiple cytokines upon T cell receptor (TCR) engagement and modulate many immune-mediated conditions. The purpose of this study was to examine the role of these cells in the development of autoimmune disease in genetically lupus-prone (NZBxNZW)F1 (BWF1) mice. METHODS The CD1d1-null genotype was crossed onto the NZB and NZW backgrounds to establish CD1d1-knockout (CD1d0) BWF1 mice. CD1d0 mice and their wild-type littermates were monitored for the development of nephritis and assessed for cytokine responses to CD1d-restricted glycolipid alpha-galactosylceramide (alphaGalCer), anti-CD3 antibody, and concanavalin A (Con A). Thymus and spleen cells were stained with CD1d tetramers that had been loaded with alphaGalCer or its analog PBS-57 to detect iNKT cells, and the cells were compared between BWF1 mice and class II major histocompatibility complex-matched nonautoimmune strains, including BALB/c, (BALB/cxNZW)F1 (CWF1), and NZW. RESULTS CD1d0 BWF1 mice had more severe nephritis than did their wild-type littermates. Although iNKT cells and iNKT cell responses were absent in CD1d0 BWF1 mice, the CD1d0 mice continued to have significant numbers of interferon-gamma-producing NKT-like (CD1d-independent TCRbeta+,NK1.1+ and/or DX5+) cells. CD1d deficiency also influenced cytokine responses by conventional T cells: upon in vitro stimulation of splenocytes with Con A or anti-CD3, type 2 cytokine levels were reduced, whereas type 1 cytokine levels were increased or unchanged in CD1d0 mice as compared with their wild-type littermates. Additionally, numbers of thymic iNKT cells were lower in young wild-type BWF1 mice than in nonautoimmune strains. CONCLUSION Germline deletion of CD1d exacerbates lupus in BWF1 mice. This finding, together with reduced thymic iNKT cells in young BWF1 mice as compared with nonautoimmune strains, implies a regulatory role of CD1d and iNKT cells during the development of lupus.
Collapse
MESH Headings
- Animals
- Antigens, CD1/genetics
- Antigens, CD1/immunology
- Antigens, CD1d
- Cells, Cultured
- Disease Models, Animal
- Galactosylceramides/pharmacology
- Gene Silencing
- Genetic Predisposition to Disease
- Interferon-gamma/metabolism
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Lupus Erythematosus, Systemic/genetics
- Lupus Nephritis/genetics
- Lupus Nephritis/immunology
- Lupus Nephritis/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred NZB
- Mice, Knockout
- Spleen/drug effects
- Spleen/metabolism
- Spleen/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- Thymus Gland/metabolism
- Thymus Gland/pathology
Collapse
Affiliation(s)
- Jun-Qi Yang
- Jun-Qi Yang, PhD, Hongzhu Liu, MD: University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Xiangshu Wen
- Xiangshu Wen, PhD, Gbolahan Folayan, BS, Xin Dong, PhD, Min Zhou, MD: David Geffen School of Medicine, University of California, Los Angeles
| | - Hongzhu Liu
- Jun-Qi Yang, PhD, Hongzhu Liu, MD: University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Gbolahan Folayan
- Xiangshu Wen, PhD, Gbolahan Folayan, BS, Xin Dong, PhD, Min Zhou, MD: David Geffen School of Medicine, University of California, Los Angeles
| | - Xin Dong
- Xiangshu Wen, PhD, Gbolahan Folayan, BS, Xin Dong, PhD, Min Zhou, MD: David Geffen School of Medicine, University of California, Los Angeles
| | - Min Zhou
- Xiangshu Wen, PhD, Gbolahan Folayan, BS, Xin Dong, PhD, Min Zhou, MD: David Geffen School of Medicine, University of California, Los Angeles
| | - Luc Van Kaer
- Luc Van Kaer, PhD: Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ram Raj Singh
- Ram Raj Singh, MD: Jonsson Comprehensive Cancer Center, and David Geffen School of Medicine, University of California, Los Angeles
| |
Collapse
|
34
|
Sireci G, Russo D, Dieli F, Porcelli SA, Taniguchi M, La Manna MP, Di Liberto D, Scarpa F, Salerno A. Immunoregulatory role of Jalpha281 T cells in aged mice developing lupus-like nephritis. Eur J Immunol 2007; 37:425-33. [PMID: 17273990 DOI: 10.1002/eji.200636695] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the emergence of autoreactive T cells. Humans and mice with SLE have reduced numbers of CD1d-restricted invariant natural killer T (iNKT) cells, suggesting a key role for these cells in its immunopathogenesis. This subset uses an invariant TCR constituted by Valpha14 Jalpha281 chains paired with some Vbeta domains. The regulatory role for iNKT cells in non-autoimmune mice was suggested by our previous results showing that aged Jalpha281 knockout (KO) mice produce anti-dsDNA. Here we show that old Jalpha281 KO mice have proteinuria and antibodies against dsDNA and cardiolipin. Histological analysis of Jalpha281 KO mice revealed glomeruli damage and deposition of C3c and IgG, mainly of the IgG3 subclass. In spleens of aged Jalpha281 KO mice there is an increase of activated marginal zone B cells. The evolution of lesions may depend on the age-associated increase of autoantibodies production, preferentially IgG3, mainly secreted by marginal zone B cells. Our results provide the first evidence of a lupus-like syndrome in non-autoimmune mice, supporting an age-related immunoregulatory role of Jalpha281+ cells, probably associated with the activation of marginal zone B cells.
Collapse
Affiliation(s)
- Guido Sireci
- Dipartimento di Biopatologia e Metodologie Biomediche, Università di Palermo, Palermo, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Deshmukh US, Bagavant H, Sim D, Pidiyar V, Fu SM. A SmD Peptide Induces Better Antibody Responses to Other Proteins within the Small Nuclear Ribonucleoprotein Complex than to SmD Protein via Intermolecular Epitope Spreading. THE JOURNAL OF IMMUNOLOGY 2007; 178:2565-71. [PMID: 17277166 DOI: 10.4049/jimmunol.178.4.2565] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Autoantibody response against the small nuclear ribonucleoprotein (snRNP) complex is a characteristic feature of systemic lupus erythematosus. The current investigation was undertaken to determine whether activation of SmD-reactive T cells by synthetic peptides harboring T cell epitopes can initiate a B cell epitope spreading cascade within the snRNP complex. T cell epitopes on SmD were mapped in A/J mice and were localized to three regions on SmD, within aa 26-55, 52-69, and 86-115. Immunization with synthetic peptides SmD(31-45), SmD(52-66), and SmD(91-110) induced T and B cell responses to the peptides, with SmD(31-45) inducing the strongest response. However, only SmD(52-66) immunization induced T cells capable of reacting with SmD. Analysis of sera by immunoprecipitation assays showed that intermolecular B cell epitope spreading to U1RNA-associated A ribonucleoprotein and SmB was consistently observed only in the SmD(52-66)-immunized mice. Surprisingly, in these mice, Ab responses to SmD were at low levels and transient. In addition, the sera did not react with other regions on SmD, indicating a lack of intramolecular B cell epitope spreading within SmD. Our study demonstrates that T cell responses to dominant epitope on a protein within a multiantigenic complex are capable of inducing B cell responses to other proteins within the complex. This effect can happen without generating a good Ab response to the protein from which the T epitope was derived. Thus caution must be taken in the identification of Ags responsible for initiating autoimmune responses based solely on serological analysis of patients and animals with systemic autoimmune disorders.
Collapse
Affiliation(s)
- Umesh S Deshmukh
- Specialized Center of Research on Systemic Lupus Erythematosus, Division of Rheumatology and Immunology, Department of Internal Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | |
Collapse
|
36
|
Abstract
Naturally occurring, CD4+ CD25+ regulatory T cells that are exported from the thymus early in life play an important role in controlling organ-specific autoimmune diseases, but they may not be critical for suppressing systemic autoimmunity in lupus. On the other hand, lupus-prone subjects appear to be deficient in generation of adaptive T-regulatory cells that can be induced by various means. We review autoantigen-specific therapeutic approaches that induce such regulatory T cells. Of particular interest are TGF-ss producing CD4+ CD25+ and CD8+ regulatory T cells that are induced by low dose tolerance therapy of lupus-prone mice with nucleosomal histone peptide epitopes, administered subcutaneously in subnanomolar doses. These regulatory T cells are not only efficient in suppressing autoantigen recognition and autoantibody production, but they also inhibit migration/accumulation of pathogenic autoimmune cells in the target organ, such as the kidneys of mice prone to develop lupus nephritis. We discuss why and under what conditions such therapeutic approaches would be beneficial in lupus patients and lupus-prone subjects.
Collapse
Affiliation(s)
- Hee-Kap Kang
- Division of Rheumatology, Departments of Medicine and Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | |
Collapse
|
37
|
Hahn BH, Ebling F, Singh RR, Singh RP, Karpouzas G, La Cava A. Cellular and molecular mechanisms of regulation of autoantibody production in lupus. Ann N Y Acad Sci 2006; 1051:433-41. [PMID: 16126985 PMCID: PMC2291525 DOI: 10.1196/annals.1361.085] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The hyperactive interaction between helper T cells and autoimmune B cells in individuals predisposed to systemic lupus erythematosus (SLE) can be interrupted by induction of regulatory and suppressor T cells. Using two strategies-high dose tolerance to an immunoglobulin-derived peptide, and minigene vaccination with DNA encoding T cell epitopes presented by MHC class I molecules-our group has induced at least three types of regulatory/suppressive T cells. They include CD8+ T cells that suppress helper T cells by cytokine secretion, CD8+ T suppressors that kill B cells making anti-DNA antibodies, and peptide-binding CD4+CD25+ regulatory T cells that suppress B cells by direct cell contact. Each of these lymphocyte subsets suppresses anti-DNA antibody production and delays the onset of nephritis in BWF1 lupus-prone mice. Patients with SLE have amino acid sequences similar to those from murine anti-DNA antibodies used in these studies, and at similar locations in the VH regions of anti-DNA immunoglobulins. Therefore, strategies described here might ultimately be useful in therapy of the human disease.
Collapse
Affiliation(s)
- Bevra H Hahn
- Division of Rheumatology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, CA 91436, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Hahn BH, Singh RP, La Cava A, Ebling FM. Tolerogenic treatment of lupus mice with consensus peptide induces Foxp3-expressing, apoptosis-resistant, TGFbeta-secreting CD8+ T cell suppressors. THE JOURNAL OF IMMUNOLOGY 2006; 175:7728-37. [PMID: 16301683 DOI: 10.4049/jimmunol.175.11.7728] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Lupus-prone (NZB x NZW)F1 mice spontaneously develop elevated titers of anti-DNA Abs that contain T cell determinants in their V(H) regions. We have previously shown that tolerization with an artificial peptide based on these T cell determinants (pConsensus (pCons)) can block production of anti-DNA Abs and prolong survival of the mice. In this study, we show that this protection depends in part on the generation of peripheral TGFbeta- and Foxp3-expressing inhibitory CD8+ (Ti) cells. These CD8+ Ti cells suppress anti-DNA IgG production both in vitro and in vivo and require up-regulated expression of both Foxp3 and TGFbeta to exert their suppressive function, as indicated by microarray analyses, small interfering RNA inhibition studies, and blocking experiments. Additionally, CD8+ Ti cells from pCons-tolerized mice were longer-lived suppressors that up-regulated expression of Bcl-2 and were more resistant to apoptosis than similar cells from naive mice. These data indicate that clinical suppression of autoimmunity after administration of pCons depends in part on the generation of CD8+ Ti cells that suppress secretion of anti-DNA Ig using mechanisms that include Foxp3, TGFbeta, and resistance to apoptosis.
Collapse
Affiliation(s)
- Bevra H Hahn
- Department of Medicine, Division of Rheumatology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | | | | | | |
Collapse
|
39
|
Guo W, Smith D, Guth A, Aviszus K, Wysocki LJ. T cell tolerance to germline-encoded antibody sequences in a lupus-prone mouse. THE JOURNAL OF IMMUNOLOGY 2005; 175:2184-90. [PMID: 16081785 DOI: 10.4049/jimmunol.175.4.2184] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The BCR V region has been implicated as a potential avenue of T cell help for autoreactive B cells in systemic lupus erythematosus. In principle, either germline-encoded or somatically generated sequences could function as targets of such help. Preceding studies have indicated that class II MHC-restricted T cells in normal mice attain a state tolerance to germline-encoded Ab diversity. In this study, we tested whether this tolerance is intact in systemic lupus erythematosus-prone (New Zealand Black x SWR)F1 mice (SNF1). Using a hybridoma sampling approach, we found that SNF1 T cells were tolerant to germline-encoded Ab sequences. Specifically, they were tolerant to germline-encoded sequences derived from a lupus anti-chromatin Ab that arose spontaneously in this strain. This was true both for diseased and prediseased mice. Thus, there does not appear to be a global defect in T cell tolerance to Ab V regions in this autoimmune-prone strain either before or during autoimmune disease.
Collapse
Affiliation(s)
- Wenzhong Guo
- Integrated Department of Immunology, National Jewish Medical and Research Center, and University of Colorado Health Science Center, Denver, CO 80206, USA
| | | | | | | | | |
Collapse
|
40
|
Singh RR. SLE: translating lessons from model systems to human disease. Trends Immunol 2005; 26:572-9. [PMID: 16153890 PMCID: PMC2291517 DOI: 10.1016/j.it.2005.08.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Revised: 08/15/2005] [Accepted: 08/25/2005] [Indexed: 11/22/2022]
Abstract
Systemic lupus erythematosus (SLE, lupus) results from immune-mediated damage to multiple organs. Its pathogenesis should be viewed as a series of steps, beginning with impaired immune regulation that permits self-reactive T-B-cell activation, which results in the production of autoantibodies. Activated T and B cells then infiltrate tissues, which along with autoantibody and immune complex deposition, triggering local events that ultimately cause organ damage. Although improved understanding of early autoimmune events might open up avenues for disease prevention, future investigations must focus on the mechanisms of end-organ damage in model systems and how to translate this knowledge into human disease. Understanding the mechanisms of each pathogenetic step would provide a rational basis for the development of disease stage-specific diagnostic markers and treatments.
Collapse
Affiliation(s)
- Ram Raj Singh
- Autoimmunity and Tolerance Lab, Div. of Rheumatology, Dept. of Medicine, Geffen School of Medicine, University of California-Los Angeles, Rm. 32-59 Rehabilitation Center, 1000 Veteran Avenue, Los Angeles, CA 90095-1670, USA.
| |
Collapse
|
41
|
Voynova EN, Tchorbanov AI, Todorov TA, Vassilev TL. Breaking of tolerance to native DNA in nonautoimmune mice by immunization with natural protein/DNA complexes. Lupus 2005; 14:543-50. [PMID: 16130511 DOI: 10.1191/0961203305lu2165oa] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A major event in the pathogenesis of systemic lupus erythematosus (SLE) is the breaking of tolerance to native DNA and the appearance of IgG anti-double-stranded (ds) DNA antibodies. The mechanisms of the losing of tolerance are not well understood. Continuous efforts have been made in the past to induce anti-native DNA IgG autoantibodies in non-autoimmune animals but the relevance of the approaches used to what happens in spontaneous disease is unclear. We succeeded in breaking tolerance to native DNA in nonautoimmune-prone BALB/c mice by immunization with natural DNA/protein complexes. These complexes included nucleosomes, crude commercial histone and nucleohistone preparations. The anti-dsDNA IgG response lasted for more than an year. IgG deposition in the kidneys of the animals was repeatedly shown. As DNA-specific B cells behave in many ways as non-autoreactive B cells, we suggest that the activity of the self-reactive B lymphocytes could be selectively inhibited in a way that mimics a physiological mechanism controlling the magnitude and duration of the IgG antibody response to foreign antigens.
Collapse
Affiliation(s)
- E N Voynova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | | | | |
Collapse
|
42
|
Kang HK, Michaels MA, Berner BR, Datta SK. Very low-dose tolerance with nucleosomal peptides controls lupus and induces potent regulatory T cell subsets. THE JOURNAL OF IMMUNOLOGY 2005; 174:3247-55. [PMID: 15749855 DOI: 10.4049/jimmunol.174.6.3247] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We induced very low-dose tolerance by injecting lupus prone (SWR x NZB)F1 (SNF1) mice with 1 mug nucleosomal histone peptide autoepitopes s.c. every 2 wk. The subnanomolar peptide therapy diminished autoantibody levels and prolonged life span by delaying nephritis, especially by reducing inflammatory cell reaction and infiltration in kidneys. H4(71-94) was the most effective autoepitope. Low-dose tolerance therapy induced CD8+, as well as CD4+ CD25+ regulatory T (Treg) cell subsets containing autoantigen-specific cells. These adaptive Treg cells suppressed IFN-gamma responses of pathogenic lupus T cells to nucleosomal epitopes at up to a 1:100 ratio and reduced autoantibody production up to 90-100% by inhibiting nucleosome-stimulated T cell help to nuclear autoantigen-specific B cells. Both CD4+ CD25+ and CD8+ Treg cells produced and required TGF-beta1 for immunosuppression, and were effective in suppressing lupus autoimmunity upon adoptive transfer in vivo. The CD4+ CD25+ T cells were partially cell contact dependent, but CD8+ T cells were contact independent. Thus, low-dose tolerance with highly conserved histone autoepitopes repairs a regulatory defect in systemic lupus erythematosus by generating long-lasting, TGF-beta-producing Treg cells, without causing allergic/anaphylactic reactions or generalized immunosuppression.
Collapse
Affiliation(s)
- Hee-Kap Kang
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
43
|
Tao D, Shangwu L, Qun W, Yan L, Wei J, Junyan L, Feili G, Boquan J, Jinquan T. CD226 expression deficiency causes high sensitivity to apoptosis in NK T cells from patients with systemic lupus erythematosus. THE JOURNAL OF IMMUNOLOGY 2005; 174:1281-90. [PMID: 15661884 DOI: 10.4049/jimmunol.174.3.1281] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Humans and mice with systemic lupus erythematosus (SLE) and related autoimmune diseases have reduced numbers of NK T cells. An association between NK T cell deficiency and autoimmune disease has been identified. However, the mechanisms for reduction of NK T cell number in patients with SLE are unknown. In the present study we report that NK T cells from active SLE patients are highly sensitive to anti-CD95-induced apoptosis compared with those from normal subjects and inactive SLE patients. CD226 expression is deficient on NK T cells from active SLE patients. The expression of one antiapoptotic member protein, survivin, is found to be selectively deficient in freshly isolated NK T cells from active SLE patients. CD226 preactivation significantly up-regulates survivin expression and activation, which can rescue active SLE NK T cells from anti-CD95-induced apoptosis. In transfected COS7 cells, we confirm that anti-CD95-mediated death signals are inhibited by activation of the CD226 pathway through stabilization of caspase-8 and caspase-3 and through activation of survivin. We therefore conclude that deficient expression of CD226 and survivin in NK T cells from active SLE is a molecular base of high sensitivity of the cells to anti-CD95-induced apoptosis. These observations offer a potential explanation for high apoptotic sensitivity of NK T cells from active SLE, and provide a new insight into the mechanism of reduction of NK T cell number in SLE and understanding the association between NK T cell deficiency and autoimmune diseases.
Collapse
MESH Headings
- Adolescent
- Adult
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/physiology
- Apoptosis/drug effects
- Apoptosis/genetics
- Apoptosis/immunology
- COS Cells
- Chlorocebus aethiops
- Down-Regulation/drug effects
- Down-Regulation/genetics
- Down-Regulation/immunology
- Female
- Gene Silencing/immunology
- Humans
- Inhibitor of Apoptosis Proteins
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/pathology
- Lupus Erythematosus, Systemic/drug therapy
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/pathology
- Male
- Microtubule-Associated Proteins/biosynthesis
- Microtubule-Associated Proteins/deficiency
- Microtubule-Associated Proteins/genetics
- Middle Aged
- Neoplasm Proteins
- Prednisone/therapeutic use
- Survivin
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
- Transfection
- fas Receptor/genetics
- fas Receptor/immunology
Collapse
Affiliation(s)
- Deng Tao
- Department of Internal Medicine, Renmin Hospital, Wuhan University, Wuhan, Peoples Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Datta SK, Zhang L, Xu L. T-helper cell intrinsic defects in lupus that break peripheral tolerance to nuclear autoantigens. J Mol Med (Berl) 2005; 83:267-78. [PMID: 15630591 DOI: 10.1007/s00109-004-0624-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2004] [Accepted: 11/16/2004] [Indexed: 12/22/2022]
Abstract
Special populations of T helper cells drive B cells to produce IgG class switched, pathogenic autoantibodies in lupus. The major source of antigenic determinants (epitopes) that trigger interactions between lupus T and B cells is nucleosomes of apoptotic cells. These epitopes can be used for antigen-specific therapy of lupus. Secondly, the autoimmune T cells of lupus are sustained because they resist anergy and activation-induced programmed cell death by markedly upregulating cyclooxygenase (COX) 2 along with the antiapoptotic molecule c-FLIP. Only certain COX-2 inhibitors block pathogenic anti-DNA autoantibody production in lupus by causing death of autoimmune T helper cells. Hence COX-2 inhibitors may work independently of their ability to block the enzymatic function of COX-2, and structural peculiarities of these select inhibitors may lead to better drug discovery and design.
Collapse
Affiliation(s)
- Syamal K Datta
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 240 East Huron St., Chicago, IL 60611, USA.
| | | | | |
Collapse
|
45
|
Riemekasten G, Langnickel D, Enghard P, Undeutsch R, Humrich J, Ebling FM, Hocher B, Humaljoki T, Neumayer H, Burmester GR, Hahn BH, Radbruch A, Hiepe F. Intravenous Injection of a D1 Protein of the Smith Proteins Postpones Murine Lupus and Induces Type 1 Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:5835-42. [PMID: 15494537 DOI: 10.4049/jimmunol.173.9.5835] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cells that recognize nucleoproteins are required for the production of anti-dsDNA Abs involved in lupus development. SmD1 83-119 (a D1 protein of the Smith (Sm) proteins, part of small nuclear ribonucleoprotein) was recently shown to provide T cell help to anti-dsDNA Abs in the NZB/NZW model of lupus. Using this model in the present study, we showed that high dose tolerance to SmD1 (600-1000 microg i.v. of SmD1(83-119) peptide/mo) delays the production of autoantibodies, postpones the onset of lupus nephritis as confirmed by histology, and prolongs survival. Tolerance to SmD1 83-119 was adoptively transferred by CD90+ T cells, which also reduce T cell help for autoreactive B cells in vitro. One week after SmD1 83-119 tolerance induction in prenephritic mice, we detected cytokine changes in cultures of CD90+ T and B220+ B cells with decreased IFN-gamma and IL-4 expression and an increase in TGFbeta. Increased frequencies of regulatory IFN-gamma+ and IL10+ CD4+ T cells were later detected. Such regulatory IL-10+/IFN-gamma+ type 1 regulatory T cells prevented autoantibody generation and anti-CD3-induced proliferation of naive T cells. In conclusion, these results indicate that SmD1 83-119 peptide may play a dominant role in the activation of helper and regulatory T cells that influence autoantibody generation and murine lupus.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Antinuclear/biosynthesis
- Antibodies, Antinuclear/metabolism
- Autoantigens/administration & dosage
- Autoantigens/immunology
- Cells, Cultured
- Cytokines/antagonists & inhibitors
- Cytokines/biosynthesis
- Dose-Response Relationship, Immunologic
- Female
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/immunology
- Immune Tolerance
- Injections, Intravenous
- Lupus Nephritis/immunology
- Lupus Nephritis/prevention & control
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred NZB
- Molecular Sequence Data
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Resting Phase, Cell Cycle/immunology
- Ribonucleoproteins, Small Nuclear/administration & dosage
- Ribonucleoproteins, Small Nuclear/immunology
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/transplantation
- Th1 Cells/cytology
- Th1 Cells/immunology
- Th1 Cells/transplantation
- snRNP Core Proteins
Collapse
Affiliation(s)
- Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
De Albuquerque DA, Saxena V, Adams DE, Boivin GP, Brunner HI, Witte DP, Singh RR. An ACE inhibitor reduces Th2 cytokines and TGF-beta1 and TGF-beta2 isoforms in murine lupus nephritis. Kidney Int 2004; 65:846-59. [PMID: 14871404 PMCID: PMC2291513 DOI: 10.1111/j.1523-1755.2004.00462.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Angiotensin-converting enzyme (ACE) inhibitors, such as captopril, are used to control hypertension. In patients and animals with primary nephropathies, these agents improve renal function more than that would be expected from their control of hypertension. Here, we examine the effects of treatment with captopril on lupus nephritis and discuss the potential mechanism(s) by which this agent exerts its renoprotective effects. METHODS Lupus-prone, NZB/NZW F1 and MRL-lpr/lpr, mice were treated with captopril or with a control antihypertensive agent, verapamil. Mice were monitored for nephritis, and their sera and tissues analyzed for cytokine and transforming growth factor-beta (TGF-beta) expression. RESULTS Captopril treatment delayed the onset of proteinuria when administered to prenephritic mice, whereas verapamil did not. Captopril treatment also retarded disease progression when given to lupus mice that had early disease, and even reversed severe proteinuria in at least some older animals with advanced disease. It reduced chronic renal lesions, but had no effect on autoantibody production. The improvement in renal disease correlated with reduced TGF-beta expression, particularly of the TGF-beta1 and TGF-beta2 isoforms, in the kidneys. Interestingly, in vivo or in vitro exposure to captopril reduced splenic levels of type 2 cytokines, interleukin (IL)-4 and IL-10, suggesting a possible role of the immune system in captopril-mediated disease modulation. CONCLUSION Since type 2 cytokines are known to promote lupus glomerulosclerosis, decreased IL-4 and IL-10 production in captopril-treated mice may be related to this agent's renoprotective effects. We argue here that ACE inhibitors not only act as selective TGF-beta inhibitors, but also as selective immunomodulators, to improve lupus nephritis.
Collapse
Affiliation(s)
- Deijanira Alves De Albuquerque
- Department of Internal Medicine, University of Cincinnati College of Medicine, Veterans Administration Medical Center and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45267-0563, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Karpouzas GA, La Cava A, Ebling FM, Singh RR, Hahn BH. Differences between CD8+ T cells in lupus-prone (NZB x NZW) F1 mice and healthy (BALB/c x NZW) F1 mice may influence autoimmunity in the lupus model. Eur J Immunol 2004; 34:2489-99. [PMID: 15307181 PMCID: PMC2291530 DOI: 10.1002/eji.200424978] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Immunization with portions of a murine antibody to DNA induced Ig peptide-reactive peripheral CD8+ inhibitory T (Ti) cells in non-autoimmune (BALB/c x NZW) F1 (CWF1) mice. Those Ti suppressed in vitro production of IgG anti-DNA by lymphocytes from MHC-matched, lupus-prone (NZB x NZW) F1 (BWF1) mice, primarily via secretion of transforming growth factor-beta (TGF-beta). However, splenic CD8+ cells from immunized BWF1 mice failed to suppress anti-DNA. Therefore, BWF1 mice were studied for defects in peripheral CD8+ T cells. The potential to suppress autoimmunity mediated by activated CD4+ helper T and B cells in BWF1 mice was assessed. As BWF1 mice aged, peripheral CD8+ T cells expanded little; fewer than 10% displayed surface markers of activation and memory. In contrast, quantities of splenic CD4+ T and B cells increased; high proportions displayed activation/memory markers. In old compared to young BWF1 mice, splenic cell secretion of two cytokines required for generation of CD8+ T effectors, IL-2 and TGF-beta, was decreased. Immunizing BWF1 mice activated peptide-reactive CD8+ T cells, but their number was decreased compared to young BWF1 or old normal mice. While peptide-reactive splenic CD8+ T cells from immunized BWF1 mice did not survive in short-term cultures, similar CD8+ T cell lines from immunized CWF1 mice expanded and on transfer into BWF1 mice delayed autoimmunity and prolonged survival. Therefore, CD8+ T cells in old BWF1 mice are impaired in expansion, acquisition of memory, secretion of cytokine, and suppression of autoimmunity. Understanding these defects might identify targets for therapy in systemic lupus erythematosus.
Collapse
Affiliation(s)
| | - Antonio La Cava
- Rheumatology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Fanny M. Ebling
- Rheumatology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Ram Raj Singh
- Autoimmunity and Tolerance Laboratory, Department Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Bevra H. Hahn
- Rheumatology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| |
Collapse
|
48
|
Munthe LA, Os A, Zangani M, Bogen B. MHC-restricted Ig V region-driven T-B lymphocyte collaboration: B cell receptor ligation facilitates switch to IgG production. THE JOURNAL OF IMMUNOLOGY 2004; 172:7476-84. [PMID: 15187126 DOI: 10.4049/jimmunol.172.12.7476] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
B cells spontaneously process their endogenous Ig and present V region peptides on their MHC class II molecules. We have here investigated whether B cells collaborate with V region-specific CD4+ T cells in vivo. By use of paired Ig L chain-transgenic and TCR-transgenic mice and cell transfer into normal hosts, we demonstrate that B cell presentation of a V(L) region peptide to CD4+ T cells results in germinal centers, plasma cells, and Ab secretion. Because the transgenic B cells have a fixed L chain but polyclonal H chains, their B cell receptor (BCR) repertoire is diverse and may bind a multitude of ligands. In a hapten-based system, BCR ligation concomitant with V region-driven T-B collaboration induced germinal center formation and an IgM --> IgG isotype switch. In the absence of BCR ligation, mainly IgM was produced. Consistent with this, prolonged V region-driven T-B collaboration resulted in high titers of IgG autoantibodies against ubiquitous self-Ags, while natural-type Abs against exotic bacteria remained IgM. Taken together, V region-driven T-B collaboration may explain induction of natural IgM Abs (absence of BCR ligation) and IgG autoantibodies (BCR ligation by autoantigen) and may be involved in the development of autoimmunity.
Collapse
Affiliation(s)
- Ludvig A Munthe
- Institute of Immunology, Rikshospitalet University Hospital, University of Oslo, N-0027 Oslo, Norway.
| | | | | | | |
Collapse
|
49
|
Crispin JC, Martínez A, Alcocer-Varela J. Quantification of regulatory T cells in patients with systemic lupus erythematosus. J Autoimmun 2004; 21:273-6. [PMID: 14599852 DOI: 10.1016/s0896-8411(03)00121-5] [Citation(s) in RCA: 307] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
CD4+ T cells that constitutively express CD25 exhibit powerful suppressive properties. Such cells have been denominated regulatory T cells (T(R)). Alterations in T(R)cells are known to cause organ-specific autoimmune disease in animal models. The aim of this work was to quantify CD4+CD25+ T cells in patients with systemic lupus erythematosus (SLE). Thirty untreated patients (ten with active disease) and ten healthy volunteers were studied. Flow cytometry was used to quantify cell populations. CD4+CD69+, CD4+CD25+ and CD4+CD25(bright) cells were considered. Peripheral blood mononuclear cell cultures were performed and supernatants collected for IL-10 and 12 measurement. CD4+CD25+ cells were significantly decreased in patients with active disease when compared to control subjects and patients without disease activity (P<0.001). CD4+CD69+ cells were increased in patients with active disease when compared to controls (P=0.041). Accordingly, CD4+CD25(bright) cells were decreased in patients with active disease compared to healthy subjects (P<0.001). IL-12 production was hampered in cells from patients during periods of active disease when compared to healthy controls and patients during remission (P<0.001). We observed a correlation between decreased T(R)number and reduced IL-12 mononuclear cell production (r=0.362, P=0.05). This work demonstrates that CD4+CD25+ T cells are decreased in patients with clinically active SLE.
Collapse
Affiliation(s)
- Jose C Crispin
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan D.F.,14000, Mexico City, Mexico
| | | | | |
Collapse
|
50
|
Snyder CM, Aviszus K, Heiser RA, Tonkin DR, Guth AM, Wysocki LJ. Activation and tolerance in CD4(+) T cells reactive to an immunoglobulin variable region. ACTA ACUST UNITED AC 2004; 200:1-11. [PMID: 15226360 PMCID: PMC2213315 DOI: 10.1084/jem.20031234] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Antibody diversity creates an immunoregulatory challenge for T cells that must cooperate with B cells, yet discriminate between self and nonself. To examine the consequences of T cell reactions to the B cell receptor (BCR), we generated a transgenic (Tg) line of mice expressing a T cell receptor (TCR) specific for a κ variable region peptide in monoclonal antibody (mAb) 36-71. The κ epitope was originally generated by a pair of somatic mutations that arose naturally during an immune response. By crossing this TCR Tg mouse with mice expressing the κ chain of mAb 36-71, we found that κ-specific T cells were centrally deleted in thymi of progeny that inherited the κTg. Maternally derived κTg antibody also induced central deletion. In marked contrast, adoptive transfer of TCR Tg T cells into κTg recipients resulted in T and B cell activation, lymphadenopathy, splenomegaly, and the production of IgG antichromatin antibodies by day 14. In most recipients, autoantibody levels increased with time, Tg T cells persisted for months, and a state of lupus nephritis developed. Despite this, Tg T cells appeared to be tolerant as assessed by severely diminished proliferative responses to the Vκ peptide. These results reveal the importance of attaining central and peripheral T cell tolerance to BCR V regions. They suggest that nondeletional forms of T tolerance in BCR-reactive T cells may be insufficient to preclude helper activity for chromatin-reactive B cells.
Collapse
Affiliation(s)
- Christopher M Snyder
- Integrated Department of Immunology, National Jewish Medical and Research Center and University of Colorado School of Medicine, 1400 Jackson St., Denver, CO 80206, USA
| | | | | | | | | | | |
Collapse
|