1
|
Radziszewska A, Peckham H, Restuadi R, Kartawinata M, Moulding D, de Gruijter NM, Robinson GA, Butt M, Deakin CT, Wilkinson MGL, Wedderburn LR, Jury EC, Rosser EC, Ciurtin C. Type I interferon and mitochondrial dysfunction are associated with dysregulated cytotoxic CD8+ T cell responses in juvenile systemic lupus erythematosus. Clin Exp Immunol 2025; 219:uxae127. [PMID: 39719886 PMCID: PMC11748002 DOI: 10.1093/cei/uxae127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/03/2024] [Accepted: 12/22/2024] [Indexed: 12/26/2024] Open
Abstract
Juvenile systemic lupus erythematosus (JSLE) is an autoimmune condition which causes significant morbidity in children and young adults and is more severe in its presentation than adult-onset SLE. While many aspects of immune dysfunction have been studied extensively in adult-onset SLE, there is limited and contradictory evidence of how cytotoxic CD8+ T cells contribute to disease pathogenesis and studies exploring cytotoxicity in JSLE are virtually non-existent. Here, we report that CD8+ T cell cytotoxic capacity is reduced in JSLE versus healthy controls, irrespective of treatment or disease activity. Transcriptomic and serum metabolomic analysis identified that this reduction in cytotoxic CD8+ T cells in JSLE was associated with upregulated type I interferon (IFN) signalling, mitochondrial dysfunction, and metabolic disturbances when compared to controls. Greater interrogation of the influence of these pathways on altered cytotoxic CD8+ T cell function demonstrated that JSLE CD8+ T cells had enlarged mitochondria and enhanced sensitivity to IFN-α leading to selective apoptosis of effector memory (EM) CD8+ T cells, which are enriched for cytotoxic mediator-expressing cells. This process ultimately contributes to the observed reduction in CD8+ T cell cytotoxicity in JSLE, reinforcing the growing evidence that mitochondrial dysfunction is a key pathogenic factor affecting multiple immune cell populations in type I IFN-driven rheumatic diseases.
Collapse
Affiliation(s)
- Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Restuadi Restuadi
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Melissa Kartawinata
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Dale Moulding
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
- Developmental Biology and Cancer Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nina M de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - George A Robinson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Maryam Butt
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Claire T Deakin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Meredyth G Ll Wilkinson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Lucy R Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Elizabeth C Jury
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| |
Collapse
|
2
|
Long H, Espinosa L, Sawalha AH. Unraveling the immunomodulatory impact of hydroxychloroquine on peripheral T cells using single-cell RNA sequencing. J Autoimmun 2024; 149:103324. [PMID: 39405653 DOI: 10.1016/j.jaut.2024.103324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/28/2024] [Accepted: 10/05/2024] [Indexed: 12/15/2024]
Abstract
Hydroxychloroquine (HCQ) is widely used in the treatment of a variety of autoimmune diseases. However, the mechanisms responsible for the immunomodulatory properties of HCQ in T cells remain unclear. Here we used single-cell RNA-sequencing to examine the effect of HCQ on T cells following in vitro stimulation. HCQ treatment led to a reduction in effector CD4+ T cells and upregulation of inhibitory genes including CTLA4 and TNFAIP3 in effector and naive CD4+ T cells, respectively. HCQ induced a significant expansion of effector CD8+ T cells, and significantly upregulated key cytotoxicity genes including GZMA, GZMB, GZMH, KLRD1, NKG7, and PRF1, as well as IFNG expression. Furthermore, HCQ treatment led to a reduction in the CD38+ CD8+ T cell subset, which is characterized by defective cytotoxicity and thought to both play a pathogenic role and increase susceptibility to infections in autoimmunity. We analyzed single-cell RNA-sequencing data in effector CD8+ T cells from lupus patients with or without HCQ treatment and confirmed upregulation of key cytotoxicity genes in patients receiving HCQ. In conclusion, this work provides additional insights into the immunomodulatory effects of HCQ and indicates that HCQ improves T cell cytotoxicity, which could explain a previously suggested protective effect of HCQ against infections in patients with autoimmune diseases.
Collapse
Affiliation(s)
- Huizhong Long
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Luis Espinosa
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
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
|
4
|
Wang K, Zhao J, Feng X, He S, Li J, Sun F, Xu Z, Yang H, Ye J, Cao L, Ye S. PD-1/PD-L1 governed cross-talk of exhausted CD8 + T and memory B cells in systemic lupus erythematosus. RMD Open 2024; 10:e003503. [PMID: 38233074 PMCID: PMC10806639 DOI: 10.1136/rmdopen-2023-003503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 01/05/2024] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Indeterminate readout of the quantitative interferon-γ release test (QFT) for Mycobacterium tuberculosis screening is a specific laboratory finding for systemic lupus erythematosus (SLE), which may be due to T-cell exhaustion and abnormal programmed death receptor 1 (PD-1)/programmed death-ligand 1 (PD-L1) signalling. METHODS We enrolled 104 patients with SLE and 225 with other rheumatic musculoskeletal diseases (RMDs) who presented to the outpatient clinic between 2020 and 2023. Twenty healthy donors served as the controls. The QFT was performed in all participants, and those with indeterminate results were compared among the groups. Immunophenotyping and functional assays were performed using blood mononuclear cells. Interferon (IFN)-γ was detected in vitro and ex vivo in patients with SLE with indeterminate or negative QFT results, before or after rituximab therapy. RESULTS 104 patients with SLE had a significantly higher rate of indeterminate QFT results was significantly higher (17.31%) than that of 225 patients with RMD (3.56%). Patients with SLE with indeterminate QFT had more active disease (SLEDAI-2K, mean 10.94 vs 4.02, p<0.0001), including a higher incidence of active nephritis (55.56% vs 29.07%). Indeterminate QFT in SLE is mainly caused by an insufficient IFN-γ response in CD8+T cells with exhausted immunophenotypes. The abnormal interaction between exhausted PD-1 high CD8+ T cells and activated PD-L1 low memory B cells in SLE can be reversed with a PD-1 agonist or increased PD-L1 expression. Rituximab treatment indirectly reversed this IFN-γ response. CONCLUSION The PD-1/PD-L1 signalling pathway, which governs the crosstalk between exhausted CD8+ T cells and activated memory B cells, is a mechanistic explanation for insufficient interferon-γ response in patients with SLE.
Collapse
Affiliation(s)
- Kaiwen Wang
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Laboratory of Rheumatology & Immunology, Jiading Branch, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiangfeng Zhao
- Laboratory of Rheumatology & Immunology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xuemei Feng
- Department of Clinical laboratory, Tibetan Medicine Hospital of Qinghai Province, Qinghai University School of Medicine, Xining, Qinghai, China
| | - Shuangjun He
- Department of Emergency, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Li
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangfang Sun
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhangling Xu
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiting Yang
- Laboratory of Rheumatology & Immunology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jiaer Ye
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liou Cao
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang Ye
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Kundura L, Cezar R, André S, Campos-Mora M, Lozano C, Vincent T, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Loubet P, Sotto A, Tran TA, Estaquier J, Corbeau P. Low perforin expression in CD8+ T lymphocytes during the acute phase of severe SARS-CoV-2 infection predicts long COVID. Front Immunol 2022; 13:1029006. [PMID: 36341327 PMCID: PMC9630742 DOI: 10.3389/fimmu.2022.1029006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/03/2022] [Indexed: 11/26/2022] Open
Abstract
T cell cytotoxicity plays a major role in antiviral immunity. Anti-SARS-CoV-2 immunity may determine acute disease severity, but also the potential persistence of symptoms (long COVID). We therefore measured the expression of perforin, a cytotoxic mediator, in T cells of patients recently hospitalized for SARS-CoV-2 infection. We recruited 54 volunteers confirmed as being SARS-CoV-2-infected by RT-PCR and admitted to Intensive Care Units (ICUs) or non-ICU, and 29 age- and sex-matched healthy controls (HCs). Amounts of intracellular perforin and granzyme-B, as well as cell surface expression of the degranulation marker CD107A were determined by flow cytometry. The levels of 15 cytokines in plasma were measured by Luminex. The frequency of perforin-positive T4 cells and T8 cells was higher in patients than in HCs (9.9 ± 10.1% versus 4.6 ± 6.4%, p = 0.006 and 46.7 ± 20.6% vs 33.3 ± 18.8%, p = 0.004, respectively). Perforin expression was neither correlated with clinical and biological markers of disease severity nor predictive of death. By contrast, the percentage of perforin-positive T8 cells in the acute phase of the disease predicted the onset of long COVID one year later. A low T8 cytotoxicity in the first days of SARS-CoV-2 infection might favor virus replication and persistence, autoimmunity, and/or reactivation of other viruses such as Epstein-Barr virus or cytomegalovirus, paving the way for long COVID. Under this hypothesis, boosting T cell cytotoxicity during the acute phase of the infection could prevent delayed sequelae.
Collapse
Affiliation(s)
- Lucy Kundura
- Institute of Human Genetics, Unité Mixte de Recherche 9002 (UMR9002), Centre National de Recherche Scientifique (CNRS) and Montpellier University, Montpellier, France
- *Correspondence: Lucy Kundura,
| | - Renaud Cezar
- Immunology Department, Nîmes University Hospital, Nîmes, France
| | - Sonia André
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1124, Université Paris Cité, Paris, France
| | - Mauricio Campos-Mora
- Institute for Regenerative Medicine & Biotherapy, Montpellier University Hospital, Montpellier, France
| | - Claire Lozano
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Thierry Vincent
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Laurent Muller
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Jean-Yves Lefrant
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Claire Roger
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Pierre-Géraud Claret
- Medical and Surgical Emergency Department, Nîmes University Hospital, Nîmes, France
| | - Sandra Duvnjak
- Gerontology Department, Nîmes University Hospital, Nîmes, France
| | - Paul Loubet
- Infectious diseases Department, Nîmes University Hospital, Nîmes, France
| | - Albert Sotto
- Infectious diseases Department, Nîmes University Hospital, Nîmes, France
| | - Tu-Ahn Tran
- Pediatrics Department, Nîmes University Hospital, Nîmes, France
| | - Jérôme Estaquier
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1124, Université Paris Cité, Paris, France
- Québec University Hospital, CHU de Québec, Laval University Research Center, Quebec City, QC, Canada
| | - Pierre Corbeau
- Institute of Human Genetics, Unité Mixte de Recherche 9002 (UMR9002), Centre National de Recherche Scientifique (CNRS) and Montpellier University, Montpellier, France
- Immunology Department, Nîmes University Hospital, Nîmes, France
| |
Collapse
|
6
|
Li H, Boulougoura A, Endo Y, Tsokos GC. Abnormalities of T cells in systemic lupus erythematosus: new insights in pathogenesis and therapeutic strategies. J Autoimmun 2022; 132:102870. [PMID: 35872102 DOI: 10.1016/j.jaut.2022.102870] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/25/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by loss of immune tolerance and sustained production of autoantibodies. Multiple and profound T cell abnormalities in SLE are intertwined with disease expression. Both numerical and functional disturbances have been reported in main CD4+ T helper cell subsets including Th1, Th2, Th17, regulatory, and follicular helper cells. SLE CD4+ T cells are known to provide help to B cells, produce excessive IL-17 but insufficient IL-2, and infiltrate tissues. In the absence of sufficient amounts of IL-2, regulatory T cells, do not function properly to constrain inflammation. A complicated series of early signaling defects and aberrant activation of kinases and phosphatases result in complex cell phenotypes by altering the metabolic profile and the epigenetic landscape. All main metabolic pathways including glycolysis, glutaminolysis and oxidative phosphorylation are altered in T cells from lupus prone mice and patients with SLE. SLE CD8+ cytotoxic T cells display reduced cytolytic activity which accounts for higher rates of infection and the sustenance of autoimmunity. Further, CD8+ T cells in the context of rheumatic diseases lose the expression of CD8, acquire IL-17+CD4-CD8- double negative T (DNT) cell phenotype and infiltrate tissues. Herein we present an update on these T cell abnormalities along with underlying mechanisms and discuss how these advances can be exploited therapeutically. Novel strategies to correct these aberrations in T cells show promise for SLE treatment.
Collapse
Affiliation(s)
- Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Afroditi Boulougoura
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yushiro Endo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Iwata S, Tanaka Y. Association of Viral Infection With the Development and Pathogenesis of Systemic Lupus Erythematosus. Front Med (Lausanne) 2022; 9:849120. [PMID: 35280878 PMCID: PMC8914279 DOI: 10.3389/fmed.2022.849120] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/03/2022] [Indexed: 12/27/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that causes multiple organ damage in women of childbearing age and has a relapsing-remitting course. SLE is caused by the interaction between genetic and environmental factors, however, its underlying triggers remain unknown. Among the environmental factors, the involvement of infections as a trigger for SLE, especially those of viral etiology, has been widely reported. Human endogenous retroviruses (HERVs) may put patients at a genetic predisposition to SLE, while the Epstein-Barr virus (EBV) may play a role as an environmental factor that triggers the development of SLE. It has been suggested that EBV-infected B-cells may become resistant to apoptosis, resulting in the activation, proliferation, and antibody production of autoreactive B-cells, which cause tissue damage in SLE. However, the interaction between the virus and immune cells, as well as the impact of the virus on the differentiation and dysfunction of immune cells, remain unclear. In this review, we focus on the relationship between the development and pathogenesis of SLE and viral infections, as well as the mechanism of SLE exacerbation via activation of immune cells, such as B-cells, based on the latest findings.
Collapse
|
8
|
Jakovcevski I, Schachner M. Perforin affects regeneration in a mouse spinal cord injury model. Int J Neurosci 2022; 132:1-12. [PMID: 32672480 DOI: 10.1080/00207454.2020.1796662] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/11/2020] [Accepted: 07/01/2020] [Indexed: 02/05/2023]
Abstract
MATERIALS AND METHODS Locomotor outcomes in perforin-deficient (Pfp-/-) mice and wild-type littermate controls were measured after severe compression injury of the lower thoracic spinal cord up to six weeks after injury. RESULTS According to both the Basso mouse scale score and single frame motion analysis, motor recovery of Pfp-/- mice was similar to wild-type controls. Interestingly, immunohistochemical analysis of cell types at six weeks after injury showed enhanced cholinergic reinnervation of spinal motor neurons caudal to the lesion site and neurofilament-positive structures at the injury site in Pfp-/- mice, whereas numbers of microglia/macrophages and astrocytes were decreased compared with controls. CONCLUSIONS We conclude that, although, loss of perforin does not change the locomotor outcome after injury, it beneficially affects diverse cellular features, such as number of axons, cholinergic synapses, astrocytes and microglia/macrophages at or caudal to the lesion site. Perforin's ability to contribute to Rag2's influence on locomotion was observed in mice doubly deficient in perforin and Rag2 which recovered better than Rag2-/- or Pfp-/- mice, suggesting that natural killer cells can cooperate with T- and B-cells in spinal cord injury.
Collapse
Affiliation(s)
- Igor Jakovcevski
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a serious autoimmune disease with a wide range of organ involvement. In addition to aberrant B-cell responses leading to autoantibody production, T-cell abnormalities are important in the induction of autoimmunity and the ensuing downstream organ damage. In this article, we present an update on how subsets of CD8+ T cells contribute to SLE pathogenesis. RECENT FINDINGS Reduced cytolytic function of CD8+ T cells not only promotes systemic autoimmunity but also accounts for the increased risk of infections. Additional information suggests that effector functions of tissue CD8+ T cells contribute to organ damage. The phenotypic changes in tissue CD8+ T cells likely arise from exposure to tissue microenvironment and crosstalk with tissue resident cells. Research on pathogenic IL-17-producing double negative T cells also suggests their origin from autoreactive CD8+ T cells, which also contribute to the induction and maintenance of systemic autoimmunity. SUMMARY Reduced CD8+ T-cell effector function illustrates their role in peripheral tolerance in the control of autoimmunity and to the increased risk of infections. Inflammatory cytokine producing double negative T cells and functional defects of regulatory CD8+ T cell both contribute to SLE pathogenesis. Further in depth research on these phenotypic changes are warranted for the development of new therapeutics for people with SLE.
Collapse
|
10
|
Hahn BH, Kono DH. Animal Models in Lupus. DUBOIS' LUPUS ERYTHEMATOSUS AND RELATED SYNDROMES 2019:164-215. [DOI: 10.1016/b978-0-323-47927-1.00014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
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
|
12
|
Azizi G, Pouyani MR, Abolhassani H, Sharifi L, dizaji MZ, Mohammadi J, Mirshafiey A, Aghamohammadi A. Cellular and molecular mechanisms of immune dysregulation and autoimmunity. Cell Immunol 2016; 310:14-26. [DOI: 10.1016/j.cellimm.2016.08.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/18/2016] [Accepted: 08/25/2016] [Indexed: 12/22/2022]
|
13
|
Boieri M, Shah P, Dressel R, Inngjerdingen M. The Role of Animal Models in the Study of Hematopoietic Stem Cell Transplantation and GvHD: A Historical Overview. Front Immunol 2016; 7:333. [PMID: 27625651 PMCID: PMC5003882 DOI: 10.3389/fimmu.2016.00333] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/18/2016] [Indexed: 12/13/2022] Open
Abstract
Bone marrow transplantation (BMT) is the only therapeutic option for many hematological malignancies, but its applicability is limited by life-threatening complications, such as graft-versus-host disease (GvHD). The last decades have seen great advances in the understanding of BMT and its related complications; in particular GvHD. Animal models are beneficial to study complex diseases, as they allow dissecting the contribution of single components in the development of the disease. Most of the current knowledge on the therapeutic mechanisms of BMT derives from studies in animal models. Parallel to BMT, the understanding of the pathophysiology of GvHD, as well as the development of new treatment regimens, has also been supported by studies in animal models. Pre-clinical experimentation is the basis for deep understanding and successful improvements of clinical applications. In this review, we retrace the history of BMT and GvHD by describing how the studies in animal models have paved the way to the many advances in the field. We also describe how animal models contributed to the understanding of GvHD pathophysiology and how they are fundamental for the discovery of new treatments.
Collapse
Affiliation(s)
- Margherita Boieri
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen , Göttingen , Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen , Göttingen , Germany
| | - Marit Inngjerdingen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
14
|
Nguyen V, Rus H, Chen C, Rus V. CTL-Promoting Effects of IL-21 Counteract Murine Lupus in the Parent→F1 Graft-versus-Host Disease Model. THE JOURNAL OF IMMUNOLOGY 2016; 196:1529-40. [DOI: 10.4049/jimmunol.1501824] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 12/10/2015] [Indexed: 01/05/2023]
|
15
|
Chen J, Ding L, Meng W, Yang J, Yan C, Xie J, Jing L, Li X, Fu Z. Vincristine-cyclophosphamide combination therapy positively affects T-cell subset distribution in systemic lupus erythematosus patients. Med Sci Monit 2015; 21:505-10. [PMID: 25683262 PMCID: PMC4335592 DOI: 10.12659/msm.893271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background This study aimed to analyze the T-cell subset distribution in systemic lupus erythematosus (SLE) patients and determine whether vincristine-cyclophosphamide combination therapy can positively affect their T-cell subset distribution to keep the disease in remission. Material/Methods Thirteen SLE patients with ‘low activity’ (SLE Disease Activity Index (SLEDAI)≤9), 17 SLE patients with ‘high activity’ (SLEDAI>9), and 15 healthy controls were recruited. SLE patients were treated with vincristine-cyclophosphamide combination therapy. CD3+, CD4+, and CD8+ T-cell percentages were analyzed by flow cytometry at baseline, 3 months, 6 months, 12–24 months, and >24 months. Results Significantly negative correlations were observed between the CD3+ and CD4+ T-cell percentages and SLEDAI scores at baseline (r=−0.471, P=0.015; r=−0.473, P=0.015, respectively). A significantly positive correlation was observed between CD4+ T-cell percentage and the complement component C3 at baseline (r=0.612, P=0.002). After 3 months of combination therapy, the CD3+ and CD4+ T-cell percentages were significantly higher than the high activity baseline (P<0.01, P<0.05, respectively). After 6 months, the CD3+, CD4+, and CD8+ T-cell percentages were all significantly higher than the high activity baseline (P<0.01, P<0.05, P<0.05, respectively). Conclusions T-cell subset distributions vary across different levels of SLE disease activity with higher CD3+ T-cell and CD4+ Th cell percentages favoring lower SLE activity. As CD3+ T-cell and CD4+ Th cell percentages negatively correlate with SLEDAI, vincristine-cyclophosphamide combination therapy appears to positively affect the T-cell subset distribution in SLE patients to keep the disease in remission by increasing their CD3+ T-cell and CD4+ Th cell percentages.
Collapse
Affiliation(s)
- Junwei Chen
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Lijuan Ding
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Wu Meng
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Jinhua Yang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Chenglan Yan
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Jianfang Xie
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Luo Jing
- Department of Rheumatology, The First Hospital of Shanxi Medical University, Shanxi, China (mainland)
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Zili Fu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| |
Collapse
|
16
|
Monson MS, Settlage RE, Mendoza KM, Rawal S, El-Nezami HS, Coulombe RA, Reed KM. Modulation of the spleen transcriptome in domestic turkey (Meleagris gallopavo) in response to aflatoxin B1 and probiotics. Immunogenetics 2015; 67:163-78. [PMID: 25597949 DOI: 10.1007/s00251-014-0825-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 12/24/2014] [Indexed: 12/19/2022]
Abstract
Poultry are highly susceptible to the immunotoxic effects of the food-borne mycotoxin aflatoxin B1 (AFB1). Exposure impairs cell-mediated and humoral immunity, limits vaccine efficacy, and increases the incidence of costly secondary infections. We investigated the molecular mechanisms of AFB1 immunotoxicity and the ability of a Lactobacillus-based probiotic to protect against aflatoxicosis in the domestic turkey (Meleagris gallopavo). The spleen transcriptome was examined by RNA sequencing (RNA-seq) of 12 individuals representing four treatment groups. Sequences (6.9 Gb) were de novo assembled to produce over 270,000 predicted transcripts and transcript fragments. Differential expression analysis identified 982 transcripts with statistical significance in at least one comparison between treatment groups. Transcripts with known immune functions comprised 27.6 % of significant expression changes in the AFB1-exposed group. Short exposure to AFB1 suppressed innate immune transcripts, especially from antimicrobial genes, but increased the expression of transcripts from E3 ubiquitin-protein ligase CBL-B and multiple interleukin-2 response genes. Up-regulation of transcripts from lymphotactin, granzyme A, and perforin 1 could indicate either increased cytotoxic potential or activation-induced cell death in the spleen during aflatoxicosis. Supplementation with probiotics was found to ameliorate AFB1-induced expression changes for multiple transcripts from antimicrobial and IL-2-response genes. However, probiotics had an overall suppressive effect on immune-related transcripts.
Collapse
Affiliation(s)
- Melissa S Monson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, 295 AS/VM, 1988 Fitch Ave., St. Paul, MN, 55108, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Soloviova K, Puliaiev M, Haas M, Via CS. In vivo maturation of allo-specific CD8 CTL and prevention of lupus-like graft-versus-host disease is critically dependent on T cell signaling through the TNF p75 receptor but not the TNF p55 receptor. THE JOURNAL OF IMMUNOLOGY 2013; 190:4562-72. [PMID: 23526821 DOI: 10.4049/jimmunol.1300091] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A third signal is required for maturation of effector CD8 CTL in addition to TCR and CD28 engagement. Inflammatory cytokines can provide a third signal; however, in nonpathogen settings (i.e., antitumor responses), the identity of the third signal is not clear. A useful model for in vivo CD8 CTL in the absence of exogenous pathogens is the alloantigen-driven parent-into F1 model of acute graft-versus-host disease (GVHD) characterized by a strong TNF-dependent donor antihost CD8 CTL T cell response. To determine whether TNF acts directly on donor T cells in a signal 3 manner, F1 mice received TNFR 1 (p55) knockout (KO) and/or TNFR 2 (p75) KO donor T cells. Donor p75 KO but not p55KO donor T cells failed to induce acute GVHD phenotype and instead induced a lupus-like chronic GVHD both short and long term because of quantitative and qualitative donor T cell defects, that is, reduced perforin, IFN-γ, and TNF production. Transfer of mixed or matched purified CD4 and CD8 T cells from wild type or p75KO donors demonstrated that optimal CTL maturation required p75 signaling in both CD4 and CD8 T cells. Despite defective p75KO CD4 help for CD8 CTL, p75KO CD4 help for B cells and autoimmunity was intact. These results provide a mechanism by which impaired CD8 CTL could contribute to reduced antiviral and antitumor responses and autoimmunity reported in patients receiving TNF blockers. Our results support the idea that selective p55 blockade may be beneficial by reducing inflammation without compromising CD8 CTL.
Collapse
Affiliation(s)
- Kateryna Soloviova
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814, USA
| | | | | | | |
Collapse
|
18
|
Eisenberg RA, Via CS. T cells, murine chronic graft-versus-host disease and autoimmunity. J Autoimmun 2012; 39:240-7. [PMID: 22704961 PMCID: PMC3578438 DOI: 10.1016/j.jaut.2012.05.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 05/24/2012] [Indexed: 01/06/2023]
Abstract
The chronic graft-versus-host disease (cGVHD) in mice is characterized by the production of autoantibodies and immunopathology characteristic of systemic lupus erythematosus (lupus). The basic pathogenesis involves the cognate recognition of foreign MHC class II of host B cells by alloreactive CD4 T cells from the donor. CD4 T cells of the host are also necessary for the full maturation of host B cells before the transfer of donor T cells. CD8 T cells play critical roles as well. Donor CD8 T cells that are highly cytotoxic can ablate or prevent the lupus syndrome, in part by killing recipient B cells. Host CD8 T cells can reciprocally downregulate donor CD8 T cells, and thus prevent them from suppressing the autoimmune process. Thus, when the donor inoculum contains both CD4 T cells and CD8 T cells, the resultant syndrome depends on the balance of activities of these various cell populations. For example, in one cGVHD model (DBA/2(C57BL/6xDBA/2)F1, the disease is more severe in females, as it is in several of the spontaneous mouse models of lupus, as well as in human disease. The mechanism of this female skewing of disease appears to depend on the relative inability of CD8 cells of the female host to downregulate the donor CD4 T cells that drive the autoantibody response. In general, then, the abnormal CD4 T cell help and the modulating roles of CD8 T cells seen in cGVHD parallel the participation of T cells in genetic lupus in mice and human lupus, although these spontaneous syndromes are presumably not driven by overt alloreactivity.
Collapse
Affiliation(s)
- Robert A Eisenberg
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 756 BRB II/III, 421 Curie Blvd., Philadelphia, PA 19104-6160, USA.
| | | |
Collapse
|
19
|
Garrett JPD, Fung I, Rupon J, Knight A, Mizesko M, Paessler M, Orange JS. Presentation of hemophagocytic lymphohistiocytosis due to a novel MUNC 13-4 mutation masked by partial therapeutic immunosuppression. Pediatr Rheumatol Online J 2012; 10:13. [PMID: 22554126 PMCID: PMC3503647 DOI: 10.1186/1546-0096-10-13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 04/05/2012] [Indexed: 11/10/2022] Open
Abstract
Hemophagocytic lymphohistiocytosis is a potentially fatal disease characterized by excessive macrophage and lymphocyte activity. Patients can be affected following immune activation after an oncologic, autoimmune or infectious trigger. An associated gene mutation may be found which impairs cytolytic lymphocyte function. We describe a pediatric case of hemophagocytic lymphohistiocytosis with a novel mutation of MUNC 13-4 whose diagnosis was confounded by concurrent immunosuppression. Clinical reassessment for hemophagocytic lymphohistiocytosis is necessary in persistently febrile patients with laboratory derangements in the setting of immunosuppressive agent exposure.
Collapse
Affiliation(s)
- Jackie P-D Garrett
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, 3550 Market Street, Philadelphia, PA 19104-4399, USA.
| | - Irene Fung
- Division of Allergy and Immunology, The Children’s Hospital of Philadelphia, 3550 Market Street, Philadelphia, PA 19104-4399, USA
| | - Jeremy Rupon
- Divisions of Hematology and Oncology, The Children’s Hospital of Philadelphia, 34th and Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Andrea Knight
- Division of Rheumatology, The Children’s Hospital of Philadelphia, 34th and Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Melissa Mizesko
- Division of Rheumatology, The Children’s Hospital of Philadelphia, 34th and Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Michelle Paessler
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, 34th and Civic Center Boulevard, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Jordan S Orange
- Division of Allergy and Immunology, The Children’s Hospital of Philadelphia, 3550 Market Street, Philadelphia, PA 19104-4399, USA
| |
Collapse
|
20
|
Pleiotropic targets: the problem of shared signaling circuitry in rheumatoid arthritis disease progression and protection. Future Med Chem 2012; 4:735-50. [PMID: 22530638 DOI: 10.4155/fmc.12.27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The immune response is replete with feedback control at many levels. These protective circuits are even functional within the arthritic joint, tempering disease to varying extents. An optimal therapy would inhibit autoimmune processes while maintaining protective circuitry. However, many of the cells and proteins that serve as important mediators of disease progression also play an active role in these protective circuits. The hypothesis considered in this review is that the inadvertent inhibition of protective circuitry adversely affects efficacy. Conversely, if therapeutics can be designed, which avoid inhibiting known regulatory circuits, efficacy will be improved. Understanding where these processes share signaling molecules will be crucial to the development of the next generation of therapeutics. This review discusses three well-defined signal transduction cascades; IL-2, IFNγ and TNF-α, and demonstrate within two cell types, T cells and macrophages, how these cytokines may contribute both to protection and to disease progression.
Collapse
|
21
|
Synthetic retinoid Am80 ameliorates chronic graft-versus-host disease by down-regulating Th1 and Th17. Blood 2011; 119:285-95. [PMID: 22077062 DOI: 10.1182/blood-2011-01-332478] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic GVHD (cGVHD) is a main cause of late death and morbidity after allogeneic hematopoietic cell transplantation, but its pathogenesis remains unclear. We investigated the roles of Th subsets in cGVHD with the use of a well-defined mouse model of cGVHD. In this model, development of cGVHD was associated with up-regulated Th1, Th2, and Th17 responses. Th1 and Th2 responses were up-regulated early after BM transplantation, followed by a subsequent up-regulation of Th17 cells. Significantly greater numbers of Th17 cells were infiltrated in the lung and liver from allogeneic recipients than those from syngeneic recipients. We then evaluated the roles of Th1 and Th17 in cGVHD with the use of IFN-γ-deficient and IL-17-deficient mice as donors. Infusion of IFN-γ(-/-) or IL-17(-/-) T cells attenuated cGVHD in the skin and salivary glands. Am80, a potent synthetic retinoid, regulated both Th1 and Th17 responses as well as TGF-β expression in the skin, resulting in an attenuation of cutaneous cGVHD. These results suggest that Th1 and Th17 contribute to the development of cGVHD and that targeting Th1 and Th17 may therefore represent a promising therapeutic strategy for preventing and treating cGVHD.
Collapse
|
22
|
Lage D, Pimentel VN, Soares TCB, Souza EM, Metze K, Cintra ML. Perforin and granzyme B expression in oral and cutaneous lichen planus - a comparative study. J Cutan Pathol 2011; 38:973-8. [DOI: 10.1111/j.1600-0560.2011.01781.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
23
|
Foster AD, Soloviova K, Puliaeva I, Puliaiev M, Puliaev R, Finkelman F, Via CS. Donor CD8 T cells and IFN-gamma are critical for sex-based differences in donor CD4 T cell engraftment and lupus-like phenotype in short-term chronic graft-versus-host disease mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:6238-54. [PMID: 21531893 PMCID: PMC3215299 DOI: 10.4049/jimmunol.1001074] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The transfer of unfractionated DBA/2J (DBA) splenocytes into B6D2F(1) (DBA → F(1)) mice results in greater donor CD4 T cell engraftment in females at day 14 that persists long-term and mediates greater female lupus-like renal disease. Although donor CD8 T cells have no demonstrated role in lupus pathogenesis in this model, we recently observed that depletion of donor CD8 T cells prior to transfer eliminates sex-based differences in renal disease long-term. In this study, we demonstrate that greater day 14 female donor CD4 engraftment is also critically dependent on donor CD8 T cells. Male DBA → F(1) mice exhibit stronger CD8-dependent day 8-10 graft-versus-host (GVH) and counter-regulatory host-versus-graft (HVG) responses, followed by stronger homeostatic contraction (days 10-12). The weaker day 10-12 GVH and HVG in females are followed by persistent donor T cell activation and increasing proliferation, expansion, and cytokine production from days 12 to 14. Lastly, greater female day 14 donor T cell engraftment, activation, and cytokine production were lost with in vivo IFN-γ neutralization from days 6 to 14. We conclude the following: 1) donor CD8 T cells enhance day 10 proliferation of donor CD4 T cells in both sexes; and 2) a weaker GVH/HVG in females allows prolonged survival of donor CD4 and CD8 T cells, allowing persistent activation. These results support the novel conclusion that sex-based differences in suboptimal donor CD8 CTL activation are critical for shaping sex-based differences in donor CD4 T cell engraftment at 2 wk and lupus-like disease long-term.
Collapse
Affiliation(s)
- Anthony D. Foster
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814
| | - Kateryna Soloviova
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814
| | - Irina Puliaeva
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814
| | - Maksym Puliaiev
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814
| | - Roman Puliaev
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814
| | - Fred Finkelman
- Department of Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45220
| | - Charles S. Via
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814
| |
Collapse
|
24
|
Pimentel VN, de Matos LS, Soares TCB, Adam R, Metze K, Correa MEP, de Souza CA, Cintra ML. Perforin and granzyme B involvement in oral lesions of lichen planus and chronic GVHD. J Oral Pathol Med 2011; 39:741-6. [PMID: 20618609 DOI: 10.1111/j.1600-0714.2010.00917.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Oral lesions of lichen planus and chronic graft-vs.-host disease (cGVHD) have similar clinical and histological features, but distinct etiology. Apoptosis induced by cytotoxic T lymphocyte has been proposed as a mechanism of keratinocytes death. Cytotoxicity can be mediated by granules containing granzyme B and perforin. Since common features can reflect similarities in immunological mechanisms, we studied the role of those molecules in both diseases. METHODS We analyzed 29 cases of oral lichen planus and 27 of oral cGVHD. The sections were studied on H&E, perforin and granzyme B staining. RESULTS The total means (epithelium plus connective tissue number) of the granzyme B- and perforin-positive cells were significantly higher in cGVHD than in oral lichen planus lesions (P<0.05). Also, it was found that the higher the number of perforin+ cells, the higher the number of granzyme-B+ cells in the epithelium and in the connective tissue for both groups (P < 0.05). In oral lichen planus, the number of single apoptotic bodies had a positive correlation with connective tissue granzyme immunostaining and a negative correlation with perforin (P<0.01). On the contrary, in oral cGVHD, the number of apoptotic body clusters presented a positive correlation with connective tissue perforin (P<0.01). CONCLUSIONS Our findings indicate that apoptosis in oral lichen planus seems to be correlated with granzyme B release, while in oral cGVHD, perforin seems to be more important. Although these diseases present clinical and histological similarities, subtle differences seem to exist in their pathogenetic mechanisms.
Collapse
|
25
|
Puliaeva I, Soloviova K, Puliaiev M, Lang T, Puliaev R, Via CS. Enhancement of suboptimal CD8 cytotoxic T cell effector function in vivo using antigen-specific CD80 defective T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:291-304. [PMID: 21115734 PMCID: PMC3226733 DOI: 10.4049/jimmunol.0902370] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
T cell upregulation of B7 molecules CD80 and CD86 limits T cell expansion in immunodeficient hosts; however, the relative roles of CD80 separate from CD86 on CD4 versus CD8 T cells in a normal immune system are not clear. To address this question, we used the parent-into-F1 (P→F1) murine model of graft-versus-host disease and transferred optimal and suboptimal doses of CD80 and/or CD86 knockout (KO) T cells into normal F1 hosts. Enhanced elimination of host B cells by KO T cells was observed only at suboptimal donor cell doses and was greatest for CD80 KO→F1 mice. Wild-type donor cells exhibited peak CD80 upregulation at day 10; CD80 KO donor cells exhibited greater peak (day 10) donor T cell proliferation and CD8 T cell effector CTL numbers versus wild-type→F1 mice. Fas or programmed cell death-1 upregulation was normal as was homeostatic contraction of CD80 KO donor cells from days 12-14. Mixing studies demonstrated that maximal host cell elimination was seen when both CD4 and CD8 T cells were CD80 deficient. These results indicate an important role for CD80 upregulation on Ag-activated CD4 and CD8 T cells in limiting expansion of CD8 CTL effectors as part of a normal immune response. Our results support further studies of therapeutic targeting of CD80 in conditions characterized by suboptimal CD8 effector responses.
Collapse
Affiliation(s)
- Irina Puliaeva
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814
| | - Kateryna Soloviova
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814
| | - Maksym Puliaiev
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814
| | - Thomas Lang
- Research Service, Baltimore Veterans Affairs Medical Center, University of Maryland School of Medicine, Baltimore, MD 21201
- Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Roman Puliaev
- 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
- Research Service, Baltimore Veterans Affairs Medical Center, University of Maryland School of Medicine, Baltimore, MD 21201
- Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
26
|
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
|
27
|
Foster AD, Haas M, Puliaeva I, Soloviova K, Puliaev R, Via CS. Donor CD8 T cell activation is critical for greater renal disease severity in female chronic graft-vs.-host mice and is associated with increased splenic ICOS(hi) host CD4 T cells and IL-21 expression. Clin Immunol 2010; 136:61-73. [PMID: 20451460 PMCID: PMC2898507 DOI: 10.1016/j.clim.2010.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 01/12/2010] [Indexed: 12/21/2022]
Abstract
Lupus-like renal disease in DBA/2-into-F1 (DBA --> F1) mice is driven by donor CD4 T cells and is more severe in females. Donor CD8 T cells have no known role. As expected, we observed that females receiving unfractionated DBA splenocytes (CD8 intact --> F1) exhibited greater clinical and histological severities of renal disease at 13 weeks compared to males. Surprisingly, sex-based differences in renal disease severity were lost in CD8 depleted --> F1 mice due to an improvement in females and a worsening in males. CD8 intact --> F1 female mice exhibited significantly greater donor and host effector (CD44(hi), CD62L(lo)) CD4 T cells and ICOS(hi) CD4 T follicular helper cells than males. CD8 depleted --> F1 female mice exhibited a reduction in the absolute numbers of host, but not donor CD4 Tfh cells and lost the significant increase in host CD4 effector cells vs. males. Greater female IL-21 expression, a product of Tfh cells, was seen in CD8 intact --> F1 and although reduced was still greater than male CD8 depleted --> F1 mice. Thus, donor CD8 T cells have a critical role in mediating sex-based differences in lupus renal disease severity possibly through greater host ICOS(hi) CD4 T cell involvement.
Collapse
MESH Headings
- Animals
- Antibodies, Antinuclear/blood
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- B-Lymphocytes/cytology
- B-Lymphocytes/metabolism
- Blood Urea Nitrogen
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/transplantation
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/transplantation
- Cell Count
- Cholesterol/blood
- Female
- Gene Expression/genetics
- Gene Expression/immunology
- Glomerulonephritis, Membranous/etiology
- Glomerulonephritis, Membranous/immunology
- Glomerulonephritis, Membranous/metabolism
- Glomerulonephritis, Membranous/pathology
- Graft vs Host Disease/blood
- Graft vs Host Disease/complications
- Graft vs Host Disease/immunology
- Graft vs Host Disease/pathology
- Immunoglobulin G/metabolism
- Inducible T-Cell Co-Stimulator Protein
- Interleukins/genetics
- Kidney/metabolism
- Kidney/pathology
- Kidney/ultrastructure
- Lupus Nephritis/diagnosis
- Lupus Nephritis/etiology
- Lupus Nephritis/immunology
- Lupus Nephritis/metabolism
- Lupus Nephritis/pathology
- Lymphocyte Depletion
- Male
- Mice
- Mice, Inbred DBA
- Mice, Inbred Strains
- Proteinuria/diagnosis
- Proteinuria/urine
- Serum Albumin/metabolism
- Sex Characteristics
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Helper-Inducer/metabolism
- Triglycerides/blood
- Interleukin-21
Collapse
Affiliation(s)
- Anthony D. Foster
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda MD 20814
| | - Mark Haas
- Department of Pathology & Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Irina Puliaeva
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda MD 20814
| | - Kateryna Soloviova
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda MD 20814
| | - Roman Puliaev
- 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
|
28
|
Via CS. Advances in lupus stemming from the parent-into-F1 model. Trends Immunol 2010; 31:236-45. [PMID: 20362509 PMCID: PMC2883015 DOI: 10.1016/j.it.2010.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 02/22/2010] [Accepted: 02/26/2010] [Indexed: 12/25/2022]
Abstract
The parent-into-F1 model has led to important advances in our understanding of lupus. Here, we review the work in murine lupus that elucidated the role of T cells and supported the conclusion that the parent-into-F1 model of induced lupus compares favorably with de facto gold standard spontaneous models of lupus. Then we focus on recent work in parent-into-F1 mice, which has yielded novel insights into unresolved controversies, such as the role of apoptosis in the pathogenesis of lupus and lupus in patients receiving TNF blockade. Finally, the review considers the evidence that supports a potential role for CD8 T cells, both cytotoxic and memory cells, in mediating disease remission.
Collapse
Affiliation(s)
- Charles S Via
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 21042, USA.
| |
Collapse
|
29
|
Kim J, Park S, Kim HA, Jung D, Kim HJ, Choi HJ, Cho HR, Kwon B. Roles of Host Nonhematopoietic Cells in Autoimmunity and Donor Cell Engraftment in Graft-versus-host Disease. Immune Netw 2010; 10:46-54. [PMID: 20532124 PMCID: PMC2881427 DOI: 10.4110/in.2010.10.2.46] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 03/25/2010] [Accepted: 03/31/2010] [Indexed: 02/04/2023] Open
Abstract
Background Graft-versus-host disease (GVHD) is initiated when alloreactive donor T cells are primed by host APCs to undergo clonal expansion and maturation. Since there is a controversy regarding the role of nonhematopoietic cells in GVHD, we wanted to investigate the influence of MHC disparity on nonhematopoietic cells on the pathogenesis of GVHD in the MHC-haplomismatched C57BL/6 (H-2b) or DBA/2 (H-2d)→unirradiated (C57BL/6×DBA/2) F1(BDF1; H-2b/d) murine model of acute GVHD (aGVHD) or chronic GVHD (cGVHD). Methods We generated (BDF1→C57BL/6), (BDF1→DBA/2), and (BDF1→BDF1) chimeras and examined GVHD-related parameters and donor cell engraftment in those chimeras. Results Using this experimental system, we found that 1) severe aGVHD across MHC Ag barrier depends on the expression of nonhematopoietically rather than hematopoietically derived alloAgs for maximal GVHD manifestations; 2) host APCs were sufficient to break B cell tolerance to self molecules in cGVHD, whereas host APCs were insufficient to induce autoimmunity in aGVHD; 3) donor cell engraftment was greatly enhanced in the host with MHC-matched nonhematopoietic cells. Conclusion Taken together, our results provide an insight into how MHC disparity on GVHD target organs contribute to the pathogenesis of GVHD.
Collapse
Affiliation(s)
- Juyang Kim
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, Ulsan, Korea
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Koreth J, Alyea EP, Murphy WJ, Welniak LA. Proteasome inhibition and allogeneic hematopoietic stem cell transplantation: a review. Biol Blood Marrow Transplant 2010; 15:1502-12. [PMID: 19896073 DOI: 10.1016/j.bbmt.2009.07.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 07/16/2009] [Indexed: 11/17/2022]
Abstract
The proteasome and its associated ubiquitin protein modification system have proved to be an important therapeutic target in the treatment of multiple myeloma and other cancers. In addition to direct antitumor effects, proteasome inhibition also exerts strong effects on nonneoplastic immune cells. This indicates that proteasome inhibition, through the use of agents like bortezomib, could be used therapeutically to modulate immune responses. In this review we explore the emerging data, both preclinical and clinical, highlighting the importance of proteasome targeting of immunologic responses, primarily in the context of allogeneic hematopoietic stem cell transplantation (HSCT), both for the control of transplant-related toxicities like acute and chronic graft-versus-host disease (aGVHD, cGHVHD), and for improved malignant disease control after allogeneic HSCT.
Collapse
Affiliation(s)
- John Koreth
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachustts, USA
| | | | | | | |
Collapse
|
31
|
Zhang L, Bertucci AM, Ramsey-Goldman R, Burt RK, Datta SK. Regulatory T cell (Treg) subsets return in patients with refractory lupus following stem cell transplantation, and TGF-beta-producing CD8+ Treg cells are associated with immunological remission of lupus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 183:6346-58. [PMID: 19841178 PMCID: PMC2784684 DOI: 10.4049/jimmunol.0901773] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Compared with conventional drug therapy, autologous hemopoietic stem cell transplantation (HSCT) can induce very-long-term remission in refractory lupus patients. Herein, we show that in posttransplant patients, both CD4(+)CD25(high)FoxP3(+) and an unusual CD8(+)FoxP3(+) Treg subset return to levels seen in normal subjects; accompanied by almost complete inhibition of pathogenic T cell response to critical peptide autoepitopes from histones in nucleosomes, the major lupus autoantigen from apoptotic cells. In addition to a stably sustained elevation of FoxP3, posttransplant CD8 T cells also maintained markedly higher expression levels of latency-associated peptide (LAP), CD103, PD-1, PD-L1, and CTLA-4, as compared with pretransplant CD8 T cells that were identically treated by a one-time activation and rest in short-term culture. The posttransplant CD8 regulatory T cells (Treg) have autoantigen-specific and nonspecific suppressive activity, which is contact independent and predominantly TGF-beta dependent. By contrast, the pretransplant CD8 T cells have helper activity, which is cell contact dependent. Although CD4(+)CD25(high) Treg cells return during clinical remission of conventional drug-treated lupus, the posttransplant patient's CD8 Treg cells are considerably more potent, and they are absent in drug-treated patients in whom CD4 T cell autoreactivity to nucleosomal epitopes persists even during clinical remission. Therefore, unlike conventional drug therapy, hemopoietic stem cell transplantation generates a newly differentiated population of LAP(high)CD103(high) CD8(TGF-beta) Treg cells, which repairs the Treg deficiency in human lupus to maintain patients in true immunological remission.
Collapse
Affiliation(s)
- Li Zhang
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Anne M. Bertucci
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Rosalind Ramsey-Goldman
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Richard K. Burt
- Division of Immunotherapy, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Syamal K. Datta
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
32
|
Cytolytic T cells induce ceramide-rich platforms in target cell membranes to initiate graft-versus-host disease. Blood 2009; 114:3693-706. [PMID: 19666872 DOI: 10.1182/blood-2008-11-191148] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Alloreactive donor cytolytic T lymphocytes play a critical role in pathophysiology of acute graft-versus-host disease (GVHD). As GVHD progression involves tumor necrosis factor superfamily receptor activation, and as apoptotic signaling for some tumor necrosis factor superfamily receptors might involve acid sphingomyelinase (ASMase)-mediated ceramide generation, we hypothesized that ASMase deletion would ameliorate GVHD. Using clinically relevant mouse models of acute GVHD in which allogeneic bone marrow and T cells were transplanted into asmase+/+ and asmase(-/-) hosts, we identify host ASMase as critical for full-blown GVHD. Lack of host ASMase reduced the acute inflammatory phase of GVHD, attenuating cytokine storm, CD8+ T-cell proliferation/activation, and apoptosis of relevant graft-versus-host target cells (hepatocytes, intestinal, and skin cells). Organ injury was diminished in asmase(-/-) hosts, and morbidity and mortality improved at 90 days after transplantation. Resistance to cytolytic T lymphocyte-induced apoptosis was found at the target cell membrane if hepatocytes lack ASMase, as hepatocyte apoptosis required target cell ceramide generation for formation of ceramide-rich macrodomains, sites concentrating proapoptotic Fas. These studies indicate a requirement for target cell ASMase in evolution of GVHD in liver, small intestines, and skin and provide potential new targets for disease management.
Collapse
|
33
|
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
|
34
|
Kim J, Park K, Kim HJ, Kim J, Kim HA, Jung D, Kim HJ, Choi HJ, Choi SY, Seo KW, Cho HR, Kwon B. Breaking of CD8+ T cell tolerance through in vivo ligation of CD40 results in inhibition of chronic graft-versus-host disease and complete donor cell engraftment. THE JOURNAL OF IMMUNOLOGY 2008; 181:7380-9. [PMID: 18981161 DOI: 10.4049/jimmunol.181.10.7380] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the DBA/2 --> unirradiated (C57BL/6 x DBA/2)F(1) model of chronic graft-vs-host disease (cGVHD), donor CD4(+) T cells play a critical role in breaking host B cell tolerance, while donor CD8(+) T cells are rapidly removed and the remaining cells fall into anergy. Previously we have demonstrated that in vivo ligation of GITR (glucocorticoid-induced TNF receptor-related gene) can activate donor CD8(+) T cells, subsequently converting the disease pattern from cGVHD to an acute form. In this study, we investigated the effect of an agonistic mAb against CD40 on cGVHD. Treatment of anti-CD40 mAb inhibited the production of anti-DNA IgG1 autoantibody and the development of glomerulonephritis. The inhibition of cGVHD occurred because anti-CD40 mAb prevented donor CD8(+) T cell anergy such that subsequently activated donor CD8(+) T cells deleted host CD4(+) T cells and host B cells involved in autoantibody production. Additionally, functionally activated donor CD8(+) T cells induced full engraftment of donor hematopoietic cells and exhibited an increased graft-vs-leukemia effect. However, induction of acute GVHD by donor CD8(+) T cells seemed to be not so apparent. Further CTL analysis indicated that there were lower levels of donor CTL activity against host cells in mice that received anti-CD40 mAb, compared with mice that received anti-GITR mAb. Taken together, our results suggest that a different intensity of donor CTL activity is required for removal of host hematopoietic cells, including leukemia vs induction of acute GVHD.
Collapse
Affiliation(s)
- Juyang Kim
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Puliaeva I, Puliaev R, Shustov A, Haas M, Via CS. Fas expression on antigen-specific T cells has costimulatory, helper, and down-regulatory functions in vivo for cytotoxic T cell responses but not for T cell-dependent B cell responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:5912-29. [PMID: 18941180 PMCID: PMC2775525 DOI: 10.4049/jimmunol.181.9.5912] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fas-mediated apoptosis is an important contributor to contraction of Ag-driven T cell responses acting only on activated Ag-specific T cells. The effects of targeted Fas deletion on selected cell populations are well described however little is known regarding the consequences of Fas deletion on only activated Ag-specific T cells. We addressed this question using the parent-into-F(1) (P-->F(1)) model of acute or chronic (lupus-like) graft-vs-host disease (GVHD) as a model of either a CTL-mediated or T-dependent B cell-mediated response, respectively. By transferring Fas-deficient lpr donor T cells into Fas-intact F(1) hosts, the in vivo role of Ag-specific T cell Fas can be determined. Our results demonstrate a novel dichotomy of Ag-specific T cell Fas function in that: 1) Fas expression on Ag-activated T cells has costimulatory, helper, and down-regulatory roles in vivo and 2) these roles were observed only in a CTL response (acute GVHD) and not in a T-dependent B cell response (chronic GVHD). Specifically, CD4 T cell Fas expression is important for optimal CD4 initial expansion and absolutely required for help for CD8 effector CTL. Donor CD8 T cell Fas expression played an important but not exclusive role in apoptosis and down-regulation. By contrast, CD4 Fas expression played no detectable role in modulating chronic GVHD induction or disease expression. These results demonstrate a novel role for Ag-specific T cell Fas expression in in vivo CTL responses and support a review of the paradigm by which Fas deficiency accelerates lupus in MRL/lpr lupus-prone mice.
Collapse
MESH Headings
- Acute Disease
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/transplantation
- Cell Line, Tumor
- Cells, Cultured
- Crosses, Genetic
- Down-Regulation/genetics
- Down-Regulation/immunology
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Graft vs Host Disease/genetics
- Graft vs Host Disease/immunology
- Graft vs Host Disease/mortality
- Lupus Nephritis/genetics
- Lupus Nephritis/immunology
- Lupus Nephritis/pathology
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred MRL lpr
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/transplantation
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- fas Receptor/biosynthesis
- fas Receptor/genetics
- fas Receptor/physiology
Collapse
Affiliation(s)
- Irina Puliaeva
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814, USA
| | | | | | | | | |
Collapse
|
36
|
Puliaev R, Puliaeva I, Welniak L, Ryan A, Haas M, Murphy W, Via CS. CTL-promoting effects of CD40 stimulation outweigh B cell-stimulatory effects resulting in B cell elimination and disease improvement in a murine model of lupus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:47-61. [PMID: 18566369 PMCID: PMC2613003 DOI: 10.4049/jimmunol.181.1.47] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD40/CD40L signaling promotes both B cell and CTL responses in vivo, the latter being beneficial in tumor models. Because CTL may also limit autoreactive B cell expansion in lupus, we asked whether an agonist CD40 mAb would exacerbate lupus due to B cell stimulation or would improve lupus due to CTL promotion. These studies used an induced model of lupus, the parent-into-F1 model in which transfer of DBA/2 splenocytes into B6D2F1 mice induces chronic lupus-like graft-vs-host disease (GVHD). Although agonist CD40 mAb treatment of DBA-->F1 mice initially exacerbated B cell expansion, it also strongly promoted donor CD8 T cell engraftment and cytolytic activity such that by 10 days host B cells were eliminated consistent with an accelerated acute GVHD. CD40 stimulation bypassed the requirement for CD4 T cell help for CD8 CTL possibly by licensing dendritic cells (DC) as shown by the following: 1) greater initial activation of donor CD8 T cells, but not CD4 T cells; 2) earlier activation of host DC; 3) host DC expansion that was CD8 dependent and CD4 independent; and 4) induction of acute GVHD using CD4-depleted purified DBA CD8+ T cells. A single dose of CD40 mAb improved lupus-like renal disease at 12 wk, but may not suffice for longer periods consistent with a need for continuing CD8 CTL surveillance. These results demonstrate that in the setting of lupus-like CD4 T cell-driven B cell hyperactivity, CTL promotion is both feasible and beneficial and the CTL-promoting properties of CD40 stimulation outweigh the B cell-stimulatory properties.
Collapse
Affiliation(s)
- Roman Puliaev
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20814, and Research Service, Baltimore VA Medical Center, and Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, Baltimore MD 21201
| | - Irina Puliaeva
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20814, and Research Service, Baltimore VA Medical Center, and Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, Baltimore MD 21201
| | - Lisbeth Welniak
- Department of Microbiology and Immunology, University of Nevada School of Medicine, Reno, NV, 89557
| | - Abigail Ryan
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20814, and Research Service, Baltimore VA Medical Center, and Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, Baltimore MD 21201
| | - Mark Haas
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - William Murphy
- Department of Microbiology and Immunology, University of Nevada School of Medicine, Reno, NV, 89557
| | - Charles S. Via
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20814, and Research Service, Baltimore VA Medical Center, and Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, Baltimore MD 21201
| |
Collapse
|
37
|
Chu YW, Gress RE. Murine models of chronic graft-versus-host disease: insights and unresolved issues. Biol Blood Marrow Transplant 2008; 14:365-78. [PMID: 18342778 PMCID: PMC2376050 DOI: 10.1016/j.bbmt.2007.12.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Accepted: 12/02/2007] [Indexed: 11/26/2022]
Abstract
Chronic graft-versus-host-disease (cGVHD) is a major barrier to successful allogeneic hematopoietic stem cell transplantation (allo-HSCT), with highly variable clinical presentations. The pathophysiology of cGVHD remains relatively poorly understood. The utilization of murine models to study cGVHD encompasses experimental challenges distinct from those that have been successfully used to study acute GVHD (aGVHD). Nevertheless, despite these challenges, murine models of cGVHD have contributed to the understanding of cGVHD, and highlight its mechanistic complexity. In this article, insights into the pathophysiology of cGVHD obtained from murine studies are summarized in the context of their relevancy to clinical cGVHD. Despite experimental limitations, current and future models of murine cGVHD will continue to provide insights into the understanding of clinical cGVHD and provide information for new therapeutic interventions.
Collapse
Affiliation(s)
- Yu-Waye Chu
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda 20892-1360, Maryland, USA.
| | | |
Collapse
|
38
|
Abstract
Over the last 50 years, mouse models of bone marrow transplantation have provided the critical links between graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL) pathophysiology and clinical practice. The initial insight from mouse models that GVHD and GVL were T cell dependent has long been confirmed clinically. More recent translations from mouse models have included the important role of inflammatory cytokines in GVHD. Newly developed concepts relating to the ability of antigen presenting cell (APC) and T cell subsets to mediate GVHD now promise significant clinical advances. The ability to use knockout and transgenic approaches to dissect mechanisms of GVHD and GVL mean that mouse systems will continue as the predominant preclinical platform. The basic transplant approach in these models, coupled with modern "real-time" immunologic imaging of GVHD and GVL is discussed.
Collapse
Affiliation(s)
- Pavan Reddy
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, 6310 CCGC, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109-0942, USA.
| | | | | |
Collapse
|
39
|
|
40
|
Hurchla MA, Sedy JR, Murphy KM. Unexpected role of B and T lymphocyte attenuator in sustaining cell survival during chronic allostimulation. THE JOURNAL OF IMMUNOLOGY 2007; 178:6073-82. [PMID: 17475832 DOI: 10.4049/jimmunol.178.10.6073] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
B and T lymphocyte attenuator (BTLA; CD272) can deliver inhibitory signals to B and T cells upon binding its ligand herpesvirus entry mediator. Because CD28, CTLA-4, programmed death-1, and ICOS regulate the development of acute graft-vs-host disease (GVHD), we wished to assess if BTLA also played a role in this T cell-mediated response. In the nonirradiated parental-into-F1 model of acute GVHD, BTLA+/+ and BTLA-/- donor lymphocytes showed equivalent engraftment and expansion during the first week of the alloresponse. Unexpectedly, BTLA-/- donor T cells failed to sustain GVHD, showing a decline in surviving donor cell numbers beginning at day 9 and greatly reduced by day 11. Similarly, inhibition of BTLA-herpesvirus entry mediator engagement by in vivo administration of a blocking anti-BTLA Ab also caused reduced survival of donor cells. Microarray analysis revealed several genes that were differentially expressed by BTLA-/- and BTLA+/+ donor CD4+ T cells preceding the decline in BTLA-/- donor T cells. Several genes influencing Th cell polarization were differentially expressed by BTLA+/+ and BTLA-/- donor cells. Additionally, the re-expression of the IL-7Ralpha subunit that occurs in BTLA+/+ donor cells after 1 wk of in vivo allostimulation was not observed in BTLA-/- donor CD4+ cells. The striking loss of BTLA-/- T cells in this model indicates a role for BTLA activity in sustaining CD4+ T cell survival under the conditions of chronic stimulation in the nonirradiated parental-into-F1 GVHD.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- B-Lymphocyte Subsets/cytology
- B-Lymphocyte Subsets/immunology
- Cell Survival/immunology
- Chronic Disease
- Disease Models, Animal
- Graft Survival/immunology
- Graft vs Host Disease/immunology
- Graft vs Host Disease/metabolism
- Graft vs Host Disease/pathology
- Isoantigens/immunology
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred MRL lpr
- Mice, Knockout
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Immunologic/physiology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
Collapse
Affiliation(s)
- Michelle A Hurchla
- Department of Pathology and Center for Immunology, Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
41
|
Rus V, Nguyen V, Puliaev R, Puliaeva I, Zernetkina V, Luzina I, Papadimitriou JC, Via CS. T cell TRAIL promotes murine lupus by sustaining effector CD4 Th cell numbers and by inhibiting CD8 CTL activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:3962-72. [PMID: 17339497 DOI: 10.4049/jimmunol.178.6.3962] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
T cells play an essential role in driving humoral autoimmunity in lupus. Molecules such as TRAIL exhibit strong T cell modulatory effects and are up-regulated in lupus, raising the possibility that they may influence disease severity. To address this possibility, we examined the role of TRAIL expression on pathogenic T cells in an induced model of murine lupus, the parent-into-F(1) (P-->F(1)) model of chronic graft-vs-host disease (GVHD), using wild-type or TRAIL-deficient donor T cells. Results were compared with mice undergoing suppressive acute GVHD. Although chronic GVHD mice exhibited less donor T cell TRAIL up-regulation and IFN-alpha-inducible gene expression than acute GVHD mice, donor CD4(+) T cell TRAIL expression in chronic GVHD was essential for sustaining effector CD4(+) Th cell numbers, for sustaining help to B cells, and for more severe lupus-like renal disease development. Conversely, TRAIL expression on donor CD8(+) T cells had a milder, but significant down-regulatory effect on CTL effector function, affecting the perforin/granzyme pathway and not the Fas ligand pathway. These results indicate that, in this model, T cell-expressed TRAIL exacerbates lupus by the following: 1) positively regulating CD4(+) Th cell numbers, thereby sustaining T cell help for B cells, and 2) to a lesser degree by negatively regulating perforin-mediated CD8(+) CTL killing that could potentially eliminate activated autoreactive B cells.
Collapse
Affiliation(s)
- Violeta Rus
- Research Service, Department of Veterans Affairs Medical Center, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Kuroiwa T, Iwasaki T, Imado T, Sekiguchi M, Fujimoto J, Sano H. Hepatocyte growth factor prevents lupus nephritis in a murine lupus model of chronic graft-versus-host disease. Arthritis Res Ther 2007; 8:R123. [PMID: 16859527 PMCID: PMC1779408 DOI: 10.1186/ar2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2006] [Revised: 07/06/2006] [Accepted: 07/14/2006] [Indexed: 11/10/2022] Open
Abstract
Chronic graft-versus-host disease (GVHD) induced in (C57BL/6 x DBA/2) F1 (BDF1) mice by the injection of DBA/2 mouse spleen cells represents histopathological changes associated with systemic lupus erythematosus (SLE), primary biliary cirrhosis (PBC) and Sjogren's syndrome (SS), as indicated by glomerulonephritis, lymphocyte infiltration into the periportal area of the liver and salivary glands. We determined the therapeutic effect of hepatocyte growth factor (HGF) gene transfection on lupus using this chronic GVHD model. Chronic GVHD mice were injected in the gluteal muscle with either HVJ liposomes containing 8 microg of the human HGF expression vector (HGF-HVJ liposomes) or mock vector (untreated control). Gene transfer was repeated at 2-week intervals during 12 weeks. HGF gene transfection effectively prevented the proteinuria and histopathological changes associated with glomerulonephritis. While liver and salivary gland sections from untreated GVHD mice showed prominent PBC- and SS-like changes, HGF gene transfection reduced these histopathological changes. HGF gene transfection greatly reduced the number of splenic B cells, host B cell major histocompatibility complex class II expression, and serum levels of IgG and anti-DNA antibodies. IL-4 mRNA expression in the spleen, liver, and kidneys was significantly decreased by HGF gene transfection. CD28 expression on DBA/2 CD4+ T cells was decreased by the addition of recombinant HGF in vitro. Furthermore, IL-4 production by DBA/2 CD4+ T cells stimulated by irradiated BDF1 dendritic cells was significantly inhibited by the addition of recombinant HGF in vitro. These results suggest that HGF gene transfection inhibited T helper 2 immune responses and reduced lupus nephritis, autoimmune sialoadenitis, and cholangitis in chronic GVHD mice. HGF may represent a novel strategy for the treatment of SLE, SS and PBC.
Collapse
Affiliation(s)
- Takanori Kuroiwa
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Tsuyoshi Iwasaki
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Takehito Imado
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Masahiro Sekiguchi
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Jiro Fujimoto
- First Department of Surgery, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Hajime Sano
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
43
|
Voskoboinik I, Smyth MJ, Trapani JA. Perforin-mediated target-cell death and immune homeostasis. Nat Rev Immunol 2007; 6:940-52. [PMID: 17124515 DOI: 10.1038/nri1983] [Citation(s) in RCA: 448] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The granule exocytosis pathway of cytotoxic lymphocytes is crucial for immune surveillance and homeostasis. The trafficking of granule components, including the membrane-disruptive protein perforin, to the immunological synapse leads to the delivery of granule proteases (granzymes) into the target cell and its destruction through apoptosis. Several independent molecular abnormalities associated with defects of either granule trafficking or perforin function can cause cytotoxic lymphocyte dysfunction. In humans, inherited perforin mutations result in severe immune dysregulation that manifests as familial haemophagocytic lymphohistiocytosis. This Review describes recent progress in defining the structure, function, biochemistry and cell biology of perforin.
Collapse
Affiliation(s)
- Ilia Voskoboinik
- Cancer Immunology Program, Peter MacCallum Cancer Centre, St. Andrew's Place, East Melbourne, Victoria 3002, Australia.
| | | | | |
Collapse
|
44
|
Graves JE, Nunley K, Heffernan MP. Off-label uses of biologics in dermatology: Rituximab, omalizumab, infliximab, etanercept, adalimumab, efalizumab, and alefacept (Part 2 of 2). J Am Acad Dermatol 2007; 56:e55-79. [PMID: 17190618 DOI: 10.1016/j.jaad.2006.07.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Revised: 06/30/2006] [Accepted: 07/22/2006] [Indexed: 12/28/2022]
Abstract
Recently, dermatologists have witnessed a revolution in our therapeutic armamentarium with the development of several novel biologic immunomodulators. Although psoriasis remains the only condition in dermatology for which the use of biologic immunomodulators has been approved by the Food and Drug Administration, these drugs have the potential to significantly impact the treatment of several inflammatory conditions in dermatology. This article includes a review of the mechanism of action, dosing, and side-effect profile, as well as a review of the current literature on off-label uses of the CD20-positive B-cell antagonist rituximab, the IgE antagonist omalizumab, the tumor necrosis factor-alpha antagonists infliximab, etanercept, and adalimumab, and the T-cell response modifiers efalizumab and alefacept.
Collapse
Affiliation(s)
- Julia E Graves
- Division of Dermatology, Washington University, St Louis, Missouri, USA
| | | | | |
Collapse
|
45
|
Meng Y, Harlin H, O'Keefe JP, Gajewski TF. Induction of cytotoxic granules in human memory CD8+ T cell subsets requires cell cycle progression. THE JOURNAL OF IMMUNOLOGY 2006; 177:1981-7. [PMID: 16849512 DOI: 10.4049/jimmunol.177.3.1981] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Memory CD8(+) T cell responses are thought to be more effective as a result of both a higher frequency of Ag-specific clones and more rapid execution of effector functions such as granule-mediated lysis. Murine models have indicated that memory CD8(+) T cells exhibit constitutive expression of perforin and can lyse targets directly ex vivo. However, the regulated expression of cytotoxic granules in human memory CD8(+) T cell subsets has been underexplored. Using intracellular flow cytometry, we observed that only a minor fraction of CD45RA(-)CD8(+) T cells, or of CD8(+) T cells reactive to EBV-HLA2 tetramer, expressed intracellular granzyme B (GrB). Induction of GrB-containing cytotoxic granules in both CD45RA(+) and CD45RA(-) cells was achieved by stimulation with anti-CD3/anti-CD28 mAb-coated beads, required at least 3 days, occurred after several rounds of cell division, and required cell cycle progression. The strongest GrB induction was seen in the CCR7(+) subpopulations, with poorest proliferation being observed in the CD45RA(-)CCR7(-) effector-memory pool. Our results indicate that, as with naive T cells, induction of cytotoxic granules in human Ag-experienced CD8(+) T cells requires time and cell division, arguing that the main numerical advantage of a memory T cell pool is a larger frequency of CTL precursors. The fact that granule induction can be achieved through TCR and CD28 ligation has implications for restoring lytic effector function in the context of antitumor immunity.
Collapse
Affiliation(s)
- Yuru Meng
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
46
|
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
|
47
|
Rowe V, Banovic T, MacDonald KP, Kuns R, Don AL, Morris ES, Burman AC, Bofinger HM, Clouston AD, Hill GR. Host B cells produce IL-10 following TBI and attenuate acute GVHD after allogeneic bone marrow transplantation. Blood 2006; 108:2485-92. [PMID: 16788097 DOI: 10.1182/blood-2006-04-016063] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Host antigen-presenting cells (APCs) are known to be critical for the induction of graft-versus-host disease (GVHD) after allogeneic bone marrow transplantation (BMT), but the relative contribution of specific APC subsets remains unclear. We have studied the role of host B cells in GVHD by using B-cell-deficient microMT mice as BMT recipients in a model of CD4-dependent GVHD to major histocompatibility complex antigens. We demonstrate that acute GVHD is initially augmented in microMT recipients relative to wild-type recipients (mortality: 85% vs 44%, P < .01), and this is the result of an increase in donor T-cell proliferation, expansion, and inflammatory cytokine production early after BMT. Recipient B cells were depleted 28-fold at the time of BMT by total body irradiation (TBI) administered 24 hours earlier, and we demonstrate that TBI rapidly induces sustained interleukin-10 (IL-10) generation from B cells but not dendritic cells (DCs) or other cellular populations within the spleen. Finally, recipient mice in which B cells are unable to produce IL-10 due to homologous gene deletion develop more severe acute GVHD than recipient mice in which B cells are wild type. Thus, the induction of IL-10 in host B cells during conditioning attenuates experimental acute GVHD.
Collapse
Affiliation(s)
- Vanessa Rowe
- Bone Marrow Transplantation Laboratory, Queensland Institute of Medical Research, 300 Herston Rd, Herston, QLD 4006, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kim J, Choi WS, Kang H, Kim HJ, Suh JH, Sakaguchi S, Kwon B. Conversion of alloantigen-specific CD8+ T cell anergy to CD8+ T cell priming through in vivo ligation of glucocorticoid-induced TNF receptor. THE JOURNAL OF IMMUNOLOGY 2006; 176:5223-31. [PMID: 16621987 DOI: 10.4049/jimmunol.176.9.5223] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this study, we investigated the effect of an agonistic mAb (DTA-1) against glucocorticoid-induced TNF receptor (GITR) in a murine model of systemic lupus erythematosus-like chronic graft-vs-host disease (cGVHD). A single dose of DTA-1 inhibited the production of anti-DNA IgG1 autoantibody and the development of glomerulonephritis, typical symptoms of cGVHD. DTA-1-treated mice showed clinical and pathological signs of acute GVHD (aGVHD), such as lymphopenia, loss of body weight, increase of donor cell engraftment, and intestinal damage, indicating that DTA-1 shifted cGVHD toward aGVHD. The conversion of cGVHD to aGVHD occurred because DTA-1 prevented donor CD8+ T cell anergy. Functionally active donor CD8+ T cells produced high levels of IFN-gamma and had an elevated CTL activity against host Ags. In in vitro MLR, anergic responder CD8+ T cells were generated, and DTA-1 stimulated the activation of these anergic CD8+ T cells. We further confirmed in vivo that donor CD8+ T cells, but not donor CD4+ T cells, were responsible for the DTA-1-mediated conversion of cGVHD to aGVHD. These results indicate that donor CD8+ T cell anergy is a restriction factor in the development of aGVHD and that in vivo ligation of GITR prevents CD8+ T cell anergy by activating donor CD8+ T cells that otherwise become anergic. In sum, our data suggest GITR as an important costimulatory molecule regulating cGVHD vs aGVHD and as a target for therapeutic intervention in a variety of related diseases.
Collapse
Affiliation(s)
- Juyang Kim
- Immunomodulation Research Center (IRC) and Department of Biological Science, University of Ulsan, San29, Mukeo-dong, Nam-ku, Ulsan, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
49
|
Colombo AA, Rusconi C, Esposito C, Bernasconi P, Caldera D, Lazzarino M, Alessandrino EP. Nephrotic syndrome after allogeneic hematopoietic stem cell transplantation as a late complication of chronic graft-versus-host disease. Transplantation 2006; 81:1087-92. [PMID: 16641591 DOI: 10.1097/01.tp.0000209496.26639.cb] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND This study aims to determine the incidence and outcome of nephrotic syndrome in patients who underwent allogeneic stem cell transplantation in a single center. METHODS Records of 279 adult patients with hematological diseases who underwent allogeneic hematopoietic stem cell transplantation were analyzed to evaluate the incidence and outcome of nephrotic syndrome. The diagnosis of chronic graft-versus-host disease was based on clinical evidence with histological confirmation whenever possible. RESULTS Of the 279 patients, 105 with a minimum follow-up of 100 days developed chronic graft-versus-host disease: six of these had nephrotic syndrome. The cumulative incidence of nephrotic syndrome was 8% at day +1,681. Patients grafted with peripheral blood stem cells had a higher probability of developing nephrotic syndrome than did those grafted with bone marrow: 24% and 3%, respectively. The pathological diagnosis was membranous glomerulonephritis in four patients, and minimal change disease in one; the diagnosis could not be histologically confirmed in the sixth patient. All patients had extensive chronic graft-versus-host disease and were receiving treatment with cyclosporine A and steroids (four patients). Response to immunosuppressive therapy with cyclosporine A and steroids was achieved in all patients at a median time of 12 weeks after transplantation. CONCLUSION Patients with chronic graft-versus-host disease may be considered to be at risk of nephrotic syndrome: careful monitoring of renal function is advisable, particularly in patients receiving allogeneic peripheral stem cell grafts.
Collapse
Affiliation(s)
- Anna A Colombo
- Division of Hematology, IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy.
| | | | | | | | | | | | | |
Collapse
|
50
|
Perruche S, Marandin A, Kleinclauss FM, Angonin R, Fresnay S, Baron MH, Tiberghien P, Saas P. Association of mixed hematopoietic chimerism with elevated circulating autoantibodies and chronic graft-versus-host disease occurrence. Transplantation 2006; 81:573-82. [PMID: 16495806 PMCID: PMC3414374 DOI: 10.1097/01.tp.0000183878.53367.77] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Use of a reduced-intensity conditioning regimen before an allogeneic hematopoietic cell transplantation is frequently associated with an early state of mixed hematopoietic chimerism. Such a coexistence of both host and donor hematopoietic cells may influence posttransplant alloreactivity and may affect the occurrence and severity of acute and chronic graft-versus-host disease (GVHD) as well as the intensity of the graft-versus-leukemia effect. Here we evaluated the relation between chimerism state after reduced-intensity conditioning transplantation (RICT), autoantibody production, and chronic GVHD (cGVHD)-related pathology. METHODS Chimerism state, circulating anticardiolipin, and antidouble stranded DNA autoantibody (Ab) titers as well as occurrence of cGVHD-like lesions were investigated in a murine RICT model. RESULTS We observed a novel association between mixed chimerism state, high levels of pathogenic IgG autoantibodies, and subsequent development of cGVHD-like lesions. Furthermore, we found that the persistence of host B cells, but not dendritic cell origin or subset, was a factor associated with the appearance of cGVHD-like lesions. The implication of host B cells was confirmed by a host origin of autoantibodies. CONCLUSION Recipient B cell persistence may contribute to the frequency and/or severity of cGVHD after RICT.
Collapse
Affiliation(s)
- Sylvain Perruche
- Interaction hôte-greffon et ingénierie cellulaire et génique en transplantation
INSERM : U645Université de Franche-ComtéEFSIFR133BFC 1, bd Alexandre Fleming 25020 Besançon, FR
| | - Aliette Marandin
- Interaction hôte-greffon et ingénierie cellulaire et génique en transplantation
INSERM : U645Université de Franche-ComtéEFSIFR133BFC 1, bd Alexandre Fleming 25020 Besançon, FR
| | - François M. Kleinclauss
- Interaction hôte-greffon et ingénierie cellulaire et génique en transplantation
INSERM : U645Université de Franche-ComtéEFSIFR133BFC 1, bd Alexandre Fleming 25020 Besançon, FR
| | - Régis Angonin
- Service d'anatomie pathologique
CHU BesançonHôpital Jean Minjoz2, bd Alexandre Fleming 25030 Besançon, FR
| | - Stéphanie Fresnay
- Interaction hôte-greffon et ingénierie cellulaire et génique en transplantation
INSERM : U645Université de Franche-ComtéEFSIFR133BFC 1, bd Alexandre Fleming 25020 Besançon, FR
- Service d'anatomie pathologique
CHU BesançonHôpital Jean Minjoz2, bd Alexandre Fleming 25030 Besançon, FR
| | - Marie Hélène Baron
- Service de radiothérapie
CHU BesançonHôpital Jean Minjoz2, bd Alexandre Fleming 25030 Besançon, FR
| | - Pierre Tiberghien
- Interaction hôte-greffon et ingénierie cellulaire et génique en transplantation
INSERM : U645Université de Franche-ComtéEFSIFR133BFC 1, bd Alexandre Fleming 25020 Besançon, FR
| | - Philippe Saas
- Interaction hôte-greffon et ingénierie cellulaire et génique en transplantation
INSERM : U645Université de Franche-ComtéEFSIFR133BFC 1, bd Alexandre Fleming 25020 Besançon, FR
| |
Collapse
|