1
|
Zhang H, Grippin A, Sun M, Ma Y, Kim BYS, Teng L, Jiang W, Yang Z. New avenues for cancer immunotherapy: Cell-mediated drug delivery systems. J Control Release 2024; 375:712-732. [PMID: 39326499 DOI: 10.1016/j.jconrel.2024.09.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/15/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
Cancer research has become increasingly complex over the past few decades as knowledge of the heterogeneity of cancer cells, their proliferative ability, and their tumor microenvironments has become available. Although conventional therapies remain the most compelling option for cancer treatment to date, immunotherapy is a promising way to harness natural immune defenses to target and kill cancer cells. Cell-mediated drug delivery systems (CDDSs) have been an active line of research for enhancing the therapeutic efficacy and specificity of cancer immunotherapy. These systems can be tailored to different types of immune cells, allowing immune evasion and accumulation in the tumor microenvironment. By enabling the targeted delivery of therapeutic agents such as immune stimulants, cytokines, antibodies, and antigens, CDDSs have improved the survival of some patients with cancer. This review summarizes the research status of CDDSs, with a focus on their underlying mechanisms of action, biology, and clinical applications. We also discuss opportunities and challenges for implementation of CDDSs into mainstream cancer immunotherapy.
Collapse
Affiliation(s)
- Huan Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Adam Grippin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Man Sun
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yifan Ma
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
2
|
Sadeghi Shaker M, Rokni M, Mahmoudi M, Farhadi E. Ras family signaling pathway in immunopathogenesis of inflammatory rheumatic diseases. Front Immunol 2023; 14:1151246. [PMID: 37256120 PMCID: PMC10225558 DOI: 10.3389/fimmu.2023.1151246] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/26/2023] [Indexed: 06/01/2023] Open
Abstract
The Ras (rat sarcoma virus) is a GTP-binding protein that is considered one of the important members of the Ras-GTPase superfamily. The Ras involves several pathways in the cell that include proliferation, migration, survival, differentiation, and fibrosis. Abnormalities in the expression level and activation of the Ras family signaling pathway and its downstream kinases such as Raf/MEK/ERK1-2 contribute to the pathogenic mechanisms of rheumatic diseases including immune system dysregulation, inflammation, and fibrosis in systemic sclerosis (SSc); destruction and inflammation of synovial tissue in rheumatoid arthritis (RA); and autoantibody production and immune complexes formation in systemic lupus erythematosus (SLE); and enhance osteoblast differentiation and ossification during skeletal formation in ankylosing spondylitis (AS). In this review, the basic biology, signaling of Ras, and abnormalities in this pathway in rheumatic diseases including SSc, RA, AS, and SLE will be discussed.
Collapse
Affiliation(s)
- Mina Sadeghi Shaker
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Rokni
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Matsuzaka Y, Yashiro R. Immune Modulation Using Extracellular Vesicles Encapsulated with MicroRNAs as Novel Drug Delivery Systems. Int J Mol Sci 2022; 23:ijms23105658. [PMID: 35628473 PMCID: PMC9146104 DOI: 10.3390/ijms23105658] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 12/13/2022] Open
Abstract
Self-tolerance involves protection from self-reactive B and T cells via negative selection during differentiation, programmed cell death, and inhibition of regulatory T cells. The breakdown of immune tolerance triggers various autoimmune diseases, owing to a lack of distinction between self-antigens and non-self-antigens. Exosomes are non-particles that are approximately 50–130 nm in diameter. Extracellular vesicles can be used for in vivo cell-free transmission to enable intracellular delivery of proteins and nucleic acids, including microRNAs (miRNAs). miRNAs encapsulated in exosomes can regulate the molecular pathways involved in the immune response through post-transcriptional regulation. Herein, we sought to summarize and review the molecular mechanisms whereby exosomal miRNAs modulate the expression of genes involved in the immune response.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, University of Tokyo, Minato-ku 108-8639, Tokyo, Japan
- Correspondence: ; Tel.: +81-3-5449-5372
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan; or
| |
Collapse
|
4
|
Abdelati AA, Eshak NY, Donia HM, El-Girby AH. Urinary Cellular Profile as a Biomarker for Lupus Nephritis. J Clin Rheumatol 2021; 27:e469-e476. [PMID: 32976199 DOI: 10.1097/rhu.0000000000001553] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND/OBJECTIVE A search for the ideal biomarker for lupus nephritis (LN) is still underway, one that can be used for early detection and correlate with the class and activity of LN. Urine is normally devoid of leukocytes; however, it has been observed that macrophages and T lymphocytes are routinely present in the urine of LN patients and those with other proliferative renal diseases. This provides the idea for their potential use as biomarkers for proliferative LN. Here, we measured the urinary CD4+, CD8+ T lymphocytes, and CD14+ monocytes in patients with systemic lupus erythematosus (SLE) as potential biomarkers for LN. METHODS A longitudinal case-control study included 30 SLE patients with LN, 30 SLE patients without past or current LN, and 20 healthy subjects as a control group. The flow cytometric analysis was done using BD FACS Calibur multiparameter flow cytometer equipped with BD CellQuest Pro software for data analysis. RESULTS CD14+ cells were the most abundant cells in the urine of LN patients. The mean numbers of urinary CD8+, CD4+, and CD14+ cells/mL were significantly higher in patients with LN than in those without. The cell counts correlated significantly with proteinuria. Urinary CD14+ cells seem to occur in much higher counts in class IV than class III LN. CONCLUSIONS Urinary CD8+, CD4+, and CD14+ cells are highly sensitive and specific markers for detecting proliferative LN. A low CD4:CD8 ratio provides a further clue. Urinary CD14 cell counts may be a potential biomarker to differentiate between the different classes of proliferative LN.
Collapse
Affiliation(s)
- Abeer Ali Abdelati
- From the Department of Internal Medicine, Rheumatology and Clinical Immunology Unit
| | - Nouran Y Eshak
- From the Department of Internal Medicine, Rheumatology and Clinical Immunology Unit
| | - Hanaa M Donia
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Amira H El-Girby
- From the Department of Internal Medicine, Rheumatology and Clinical Immunology Unit
| |
Collapse
|
5
|
Deng Q, Luo Y, Chang C, Wu H, Ding Y, Xiao R. The Emerging Epigenetic Role of CD8+T Cells in Autoimmune Diseases: A Systematic Review. Front Immunol 2019; 10:856. [PMID: 31057561 PMCID: PMC6482221 DOI: 10.3389/fimmu.2019.00856] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/02/2019] [Indexed: 12/19/2022] Open
Abstract
Autoimmune diseases are usually complex and multifactorial, characterized by aberrant production of autoreactive immune cells and/or autoantibodies against healthy cells and tissues. However, the pathogenesis of autoimmune diseases has not been clearly elucidated. The activation, differentiation, and development of CD8+ T cells can be affected by numerous inflammatory cytokines, transcription factors, and chemokines. In recent years, epigenetic modifications have been shown to play an important role in the fate of CD8+ T cells. The discovery of these modifications that contribute to the activation or suppression of CD8+ cells has been concurrent with the increasing evidence that CD8+ T cells play a role in autoimmunity. These relationships have been studied in various autoimmune diseases, including multiple sclerosis (MS), systemic sclerosis (SSc), type 1 diabetes (T1D), Grave's disease (GD), systemic lupus erythematosus (SLE), aplastic anemia (AA), and vitiligo. In each of these diseases, genes that play a role in the proliferation or activation of CD8+ T cells have been found to be affected by epigenetic modifications. Various cytokines, transcription factors, and other regulatory molecules have been found to be differentially methylated in CD8+ T cells in autoimmune diseases. These genes are involved in T cell regulation, including interferons, interleukin (IL),tumor necrosis factor (TNF), as well as linker for activation of T cells (LAT), cytotoxic T-lymphocyte–associated antigen 4 (CTLA4), and adapter proteins. MiRNAs also play a role in the pathogenesis of these diseases and several known miRNAs that are involved in these diseases have also been shown to play a role in CD8+ regulation.
Collapse
Affiliation(s)
- Qiancheng Deng
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yangyang Luo
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Children's Hospital, Changsha, China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Haijing Wu
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Ding
- Department of Dermatology, Hainan Provincial Dermatology Disease Hospital, Haikou, China
| | - Rong Xiao
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
6
|
Kolb NA, Trevino CR, Waheed W, Sobhani F, Landry KK, Thomas AA, Hehir M. Neuromuscular complications of immune checkpoint inhibitor therapy. Muscle Nerve 2018; 58:10-22. [PMID: 29342325 DOI: 10.1002/mus.26070] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 01/10/2018] [Accepted: 01/14/2018] [Indexed: 12/31/2022]
Abstract
Immune checkpoint inhibitor (ICPI) therapy unleashes the body's natural immune system to fight cancer. ICPIs improve overall cancer survival, however, the unbridling of the immune system may induce a variety of immune-related adverse events. Neuromuscular immune complications are rare but they can be severe. Myasthenia gravis and inflammatory neuropathy are the most common neuromuscular adverse events but a variety of others including inflammatory myopathy are reported. The pathophysiologic mechanism of these autoimmune disorders may differ from that of non-ICPI-related immune diseases. Accordingly, while the optimal treatment for ICPI-related neuromuscular disorders generally follows a traditional paradigm, there are important novel considerations in selecting appropriate immunosuppressive therapy. This review presents 2 new cases, a summary of neuromuscular ICPI complications, and an approach to the diagnosis and treatment of these disorders. Muscle Nerve, 2018.
Collapse
Affiliation(s)
- Noah A Kolb
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont, USA
| | | | - Waqar Waheed
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont, USA
| | - Fatemeh Sobhani
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont, USA
| | - Kara K Landry
- Department of Medicine, University of Vermont Medical Center, 1 South Prospect Street, MS 405AR2, Burlington, Vermont, 05401, USA
| | - Alissa A Thomas
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont, USA
| | - Mike Hehir
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
7
|
Abraham Y, Gerrits B, Ludwig MG, Rebhan M, Gubser Keller C. Exploring Glucocorticoid Receptor Agonists Mechanism of Action Through Mass Cytometry and Radial Visualizations. CYTOMETRY PART B-CLINICAL CYTOMETRY 2017; 92:42-56. [DOI: 10.1002/cyto.b.21499] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 11/18/2016] [Accepted: 12/01/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Yann Abraham
- Developmental and Molecular Pathway, Computational Biology; Novartis Institute for Biomedical Research; Novartis Campus CH-4056 Basel, Switzerland
| | - Bertran Gerrits
- Developmental and Molecular Pathway, Computational Biology; Novartis Institute for Biomedical Research; Novartis Campus CH-4056 Basel, Switzerland
| | - Marie-Gabrielle Ludwig
- Developmental and Molecular Pathway Immunity; Novartis Institute for Biomedical Research; Novartis Campus CH-4056 Basel, Switzerland
| | - Michael Rebhan
- Developmental and Molecular Pathway, Computational Biology; Novartis Institute for Biomedical Research; Novartis Campus CH-4056 Basel, Switzerland
| | - Caroline Gubser Keller
- Developmental and Molecular Pathway, Computational Biology; Novartis Institute for Biomedical Research; Novartis Campus CH-4056 Basel, Switzerland
| |
Collapse
|
8
|
Sorrenti V, Marenda B, Fortinguerra S, Cecchetto C, Quartesan R, Zorzi G, Zusso M, Giusti P, Buriani A. Reference Values for a Panel of Cytokinergic and Regulatory Lymphocyte Subpopulations. Immune Netw 2016; 16:344-357. [PMID: 28035210 PMCID: PMC5195844 DOI: 10.4110/in.2016.16.6.344] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/03/2016] [Accepted: 12/09/2016] [Indexed: 12/11/2022] Open
Abstract
Lymphocyte subpopulations producing cytokines and exerting regulatory functions represent key immune elements. Given their reciprocal interdependency lymphocyte subpopulations are usually assayed as diagnostic panels, rather than single biomarkers for specialist clinical use. This retrospective analysis on lymphocyte subpopulations, analyzed over the last few years in an outpatient laboratory in Northeast Italy, contributes to the establishment of reference values for several regulatory lymphocytes currently lacking such reference ranges for the general population. Mean values and ranges in a sample of Caucasian patients (mean age 42±8,5 years), were provided for Th1, Th2, Th17, Th-reg, Tc-reg, Tc-CD57+ and B1 lymphocytes. The results are consistent with what is found in literature for the single subtypes and are: Th1 157.8±60.3/µl (7.3%±2.9); Th2 118.2±52.2/µl (5.4%±2.5); Th17 221.6±90.2/µl (10.5%±4.4); Th-reg 15.1±10.2/µl (0.7%±0.4); Tc-reg 5.8±4.7/µl (0.3%±0.2); Tc-CD57+ 103.7±114.1/µl (4.6%±4.7); B1 33.7±22.8/µl (1.5%±0.9); (Values are mean±SD). The results show that despite their variability, mean values are rather consistent in all age or sex groups and can be used as laboratory internal reference for this regulatory panel. Adding regulatory cells to lymphocyte subpopulations panels allows a more complete view of the state of the subject's immune network balance, thus improving the personalization and the "actionability" of diagnostic data in a systems medicine perspective.
Collapse
Affiliation(s)
- Vincenzo Sorrenti
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group, Padova 35100, Italy.; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35100, Italy
| | - Bruno Marenda
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group, Padova 35100, Italy
| | - Stefano Fortinguerra
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group, Padova 35100, Italy.; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35100, Italy
| | - Claudia Cecchetto
- Department of Biomedical Sciences, University of Padova, Padova 35100, Italy
| | - Roberta Quartesan
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group, Padova 35100, Italy
| | - Giulia Zorzi
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group, Padova 35100, Italy
| | - Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35100, Italy
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35100, Italy
| | - Alessandro Buriani
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group, Padova 35100, Italy.; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35100, Italy
| |
Collapse
|
9
|
Loochtan AI, Nickolich MS, Hobson-Webb LD. Myasthenia gravis associated with ipilimumab and nivolumab in the treatment of small cell lung cancer. Muscle Nerve 2015; 52:307-8. [PMID: 25759003 DOI: 10.1002/mus.24648] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Aaron I Loochtan
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| | - Myles S Nickolich
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Lisa D Hobson-Webb
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
10
|
Brown TT, Choi EYK, Thomas DG, Hristov AC, Chan MP. Comparative analysis of rosacea and cutaneous lupus erythematosus: Histopathologic features, T-cell subsets, and plasmacytoid dendritic cells. J Am Acad Dermatol 2014; 71:100-7. [DOI: 10.1016/j.jaad.2014.01.892] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/17/2014] [Accepted: 01/21/2014] [Indexed: 01/14/2023]
|
11
|
Luan YY, Yao YM, Sheng ZY. Update on the immunological pathway of negative regulation in acute insults and sepsis. J Interferon Cytokine Res 2012; 32:288-98. [PMID: 22509978 PMCID: PMC3390969 DOI: 10.1089/jir.2011.0117] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 01/16/2012] [Indexed: 12/20/2022] Open
Abstract
Sepsis with subsequent multiple organ dysfunction is a distinctly systemic inflammatory response to concealed or known infection and is a leading cause of death in intensive care units. In the initial stage of sepsis, a phase of immune activation can be evident, but a marked apoptosis-induced depletion of lymphocytes and a nonspecific anergy of immune function after severe trauma and burns might be responsible for the increased susceptibility of the host to subsequent septic complications. Recent studies indicated that negative regulation of immune function plays a pivotal role in the maintenance of peripheral homeostasis and regulation of immune responses; therefore, an understanding of the basic pathways might give rise to novel insights into the mechanisms of sepsis and immune homeostasis. This review is an attempt to provide a summary of the different pathways of negative regulation that are involved in the pathogenesis of sepsis, secondary to acute insults.
Collapse
Affiliation(s)
- Ying-yi Luan
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People's Republic of China
| | | | | |
Collapse
|
12
|
Kim JS, Cho BA, Sim JH, Shah K, Woo CM, Lee EB, Lee DS, Kang JS, Lee WJ, Park CG, Craft J, Kang I, Kim HR. IL-7Rαlow memory CD8+ T cells are significantly elevated in patients with systemic lupus erythematosus. Rheumatology (Oxford) 2012; 51:1587-94. [PMID: 22661557 DOI: 10.1093/rheumatology/kes100] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Human effector memory (EM) CD8(+) T cells include IL-7Rα(high) and IL-7Rα(low) cells with distinct cellular characteristics, including the expression of cytotoxic molecules. Both NK cells and the NK cell-associated molecule 2B4 that is expressed on CD8(+) T cells promote cytotoxicity. Here we analysed the expression of 2B4 on IL-7Rα(high) and IL-7Rα(low) EM CD8(+) T cells and its contribution to cytotoxicity. We also analysed the frequency of IL-7Rα(high) and IL-7Rα(low) EM CD8(+) T cells in patients with SLE or lupus and in healthy individuals given the potential role of cytotoxic CD8(+) T cells in the pathogenesis of lupus. METHODS We used flow cytometry to measure the expression of 2B4 on IL-7Rα(high) and IL-7Rα(low) EM CD8(+) T cells as well as the frequency of these cell populations in the peripheral blood of healthy individuals and patients with SLE. Also, 2B4-mediated cytotoxicity was quantitated in IL-7Rα(high) and IL-7Rα(low) EM CD8(+) T cells using target cells with CD48 antigen. RESULTS We found that IL-7Rα(high) EM CD8(+) T cells had higher levels of 2B4 expression compared with IL-7Rα(low) EM CD8(+) T cells. Triggering 2B4 enhanced the cytotoxic function of IL-7Rα(low) EM CD8(+) T cells against target cells. We also noticed that patients with SLE had an increased frequency of IL-7Rα(low) EM CD8(+) T cells that correlated with disease manifestation. CONCLUSION Our findings show that SLE patients have increased IL-7Rα(low) EM CD8(+) T cells, possibly contributing to tissue damage through 2B4-mediated cytotoxicity.
Collapse
Affiliation(s)
- Jung-Sik Kim
- Department of Anatomy, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Apostolidis SA, Lieberman LA, Kis-Toth K, Crispín JC, Tsokos GC. The dysregulation of cytokine networks in systemic lupus erythematosus. J Interferon Cytokine Res 2011; 31:769-79. [PMID: 21877904 PMCID: PMC3189553 DOI: 10.1089/jir.2011.0029] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 06/16/2011] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease associated with chronic immune activation and tissue damage. Organ damage in SLE results from the deposition of immune complexes and the infiltration of activated T cells into susceptible organs. Cytokines are intimately involved in every step of the SLE pathogenesis. Defective immune regulation and uncontrolled lymphocyte activation, as well as increased antigen presenting cell maturation are all influenced by cytokines. Moreover, expansion of local immune responses as well as tissue infiltration by pathogenic cells is instigated by cytokines. In this review, we describe the main cytokine abnormalities reported in SLE and discuss the mechanisms that drive their aberrant production as well as the pathogenic pathways that their presence promotes.
Collapse
Affiliation(s)
- Sokratis A Apostolidis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
14
|
Jeong JM, Kim JW, Park HJ, Song JH, Kim DH, Park CI. Molecular cloning and characterisation of the rock bream, Oplegnathus fasciatus, Fas (CD95/APO-1), and its expression analysis in response to bacterial or viral infection. RESULTS IN IMMUNOLOGY 2011; 1:11-7. [PMID: 24371547 DOI: 10.1016/j.rinim.2011.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 06/30/2011] [Accepted: 06/30/2011] [Indexed: 11/30/2022]
Abstract
Fas belongs to the tumour necrosis factor (TNF) receptor superfamily and can transmit a death signal leading to apoptosis. In the present study, we isolated the full-length cDNA for rock bream (Oplegnathus fasciatus) Fas (RbFas). The full-length RbFas cDNA was 1770 bp long and contained an open reading frame of 957 bp that encoded 319 amino acid residues with a predicted molecular mass of 35.1 kDa. The 319 amino-acid predicted RbFas sequence is homologous to other Fas sequences, contains three cysteine-rich domains and a death domain (DD) and two potential N-glycosylation sites. Expression of RbFas mRNA was detected in nine different tissues from healthy rock bream and was the highest in red blood cells. In analyses of mitogen-stimulated RbFas expression in peripheral blood leucocytes, expression of RbFas mRNA was observed between 1 and 36 h after stimulation with LPS, and 1 and 3 h stimulation with poly I:C. In the case of bacterial injection, the RbFas transcript peaked 6 h after injection in both the kidney and the spleen. Otherwise, the RbFas transcript peaked after 1 h in spleen and 6 h in kidney following injection with RSIV.
Collapse
Affiliation(s)
- Ji-Min Jeong
- Department of Marine Biology and Aquaculture, Institute of Marine Industry, College of Marine Science, Gyeongsang National University, 455, Tongyeong 650-160, Republic of Korea
| | - Ju-Won Kim
- Department of Marine Biology and Aquaculture, Institute of Marine Industry, College of Marine Science, Gyeongsang National University, 455, Tongyeong 650-160, Republic of Korea
| | - Hyoung-Jun Park
- Department of Marine Biology and Aquaculture, Institute of Marine Industry, College of Marine Science, Gyeongsang National University, 455, Tongyeong 650-160, Republic of Korea
| | - Jeong-Hun Song
- The College of Fisheries Science, Pukyong National University, Busan 608-737, Republic of Korea
| | - Do-Hyung Kim
- Fish Health Center and Department of Aqualife Medicine, Chonnam National University, Yeosu 550-749, Republic of Korea
| | - Chan-Il Park
- Department of Marine Biology and Aquaculture, Institute of Marine Industry, College of Marine Science, Gyeongsang National University, 455, Tongyeong 650-160, Republic of Korea
| |
Collapse
|
15
|
Tauzin S, Chaigne-Delalande B, Selva E, Khadra N, Daburon S, Contin-Bordes C, Blanco P, Le Seyec J, Ducret T, Counillon L, Moreau JF, Hofman P, Vacher P, Legembre P. The naturally processed CD95L elicits a c-yes/calcium/PI3K-driven cell migration pathway. PLoS Biol 2011; 9:e1001090. [PMID: 21713032 PMCID: PMC3119658 DOI: 10.1371/journal.pbio.1001090] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 05/11/2011] [Indexed: 02/06/2023] Open
Abstract
Patients affected by chronic inflammatory disorders display high amounts of soluble CD95L. This homotrimeric ligand arises from the cleavage by metalloproteases of its membrane-bound counterpart, a strong apoptotic inducer. In contrast, the naturally processed CD95L is viewed as an apoptotic antagonist competing with its membrane counterpart for binding to CD95. Recent reports pinpointed that activation of CD95 may attract myeloid and tumoral cells, which display resistance to the CD95-mediated apoptotic signal. However, all these studies were performed using chimeric CD95Ls (oligomerized forms), which behave as the membrane-bound ligand and not as the naturally processed CD95L. Herein, we examine the biological effects of the metalloprotease-cleaved CD95L on CD95-sensitive activated T-lymphocytes. We demonstrate that cleaved CD95L (cl-CD95L), found increased in sera of systemic lupus erythematosus (SLE) patients as compared to that of healthy individuals, promotes the formation of migrating pseudopods at the leading edge of which the death receptor CD95 is capped (confocal microscopy). Using different migration assays (wound healing/Boyden Chamber/endothelial transmigration), we uncover that cl-CD95L promotes cell migration through a c-yes/Ca²⁺/PI3K-driven signaling pathway, which relies on the formation of a CD95-containing complex designated the MISC for Motility-Inducing Signaling Complex. These findings revisit the role of the metalloprotease-cleaved CD95L and emphasize that the increase in cl-CD95L observed in patients affected by chronic inflammatory disorders may fuel the local or systemic tissue damage by promoting tissue-filtration of immune cells.
Collapse
Affiliation(s)
- Sébastien Tauzin
- Université de Rennes-1, Rennes, France
- IRSET/EA-4427 SeRAIC, Rennes, France
| | | | - Eric Selva
- Université de Nice Sophia antipolis, INSERM ERI21/EA 4319, Nice, France
| | - Nadine Khadra
- Université de Rennes-1, Rennes, France
- IRSET/EA-4427 SeRAIC, Rennes, France
| | - Sophie Daburon
- CNRS UMR 5164, Bordeaux, France
- Université de Bordeaux-2, Bordeaux, France
| | - Cécile Contin-Bordes
- CNRS UMR 5164, Bordeaux, France
- Université de Bordeaux-2, Bordeaux, France
- CHU Bordeaux, Bordeaux, France
| | - Patrick Blanco
- CNRS UMR 5164, Bordeaux, France
- Université de Bordeaux-2, Bordeaux, France
- CHU Bordeaux, Bordeaux, France
| | - Jacques Le Seyec
- Université de Rennes-1, Rennes, France
- IRSET/EA-4427 SeRAIC, Rennes, France
| | - Thomas Ducret
- Université de Bordeaux-2, Bordeaux, France
- INSERM U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France
| | - Laurent Counillon
- Université de Nice-Sophia Antipolis, UMR 6097 Faculté des Sciences Parc Valrose, Nice, France
| | - Jean-François Moreau
- CNRS UMR 5164, Bordeaux, France
- Université de Bordeaux-2, Bordeaux, France
- CHU Bordeaux, Bordeaux, France
| | - Paul Hofman
- Université de Nice Sophia antipolis, INSERM ERI21/EA 4319, Nice, France
- CHU de Nice et Centre de Ressources Biologiques-Tumorothèque, Nice, France
| | - Pierre Vacher
- Université de Bordeaux-2, Bordeaux, France
- INSERM U916, Institut Bergonié, Bordeaux, France
| | - Patrick Legembre
- Université de Rennes-1, Rennes, France
- IRSET/EA-4427 SeRAIC, Rennes, France
- CNRS UMR 5164, Bordeaux, France
- Université de Bordeaux-2, Bordeaux, France
| |
Collapse
|
16
|
The human cathelicidin, LL-37, induces granzyme-mediated apoptosis in cytotoxic T lymphocytes. Exp Cell Res 2010; 317:531-8. [PMID: 21134367 DOI: 10.1016/j.yexcr.2010.11.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 11/23/2010] [Accepted: 11/29/2010] [Indexed: 11/23/2022]
Abstract
LL-37 is a human cationic host defense peptide (antimicrobial peptide) belonging to the cathelicidin family of peptides. In this study, LL-37 was shown to kill stimulated CD8(+) T cells (Cytotoxic T lymphocytes; CTLs) via apoptosis, while having no cytotoxic effect on non-stimulated CD8(+) or CD4(+) T cells or stimulated CD4(+) T cells. Of interest, the CD8(+) cells were much more sensitive to LL-37 than many other cell types. LL-37 exposure resulted in DNA fragmentation, chromatin condensation, and the release of both granzyme A and granzyme B from intracellular granules. The importance of granzyme family members in the apoptosis of CTLs following LL-37 treatment was analyzed by using C57BL/6 lymphocytes obtained from mice that were homozygous for null mutations in the granzyme B gene, the granzyme A gene, or both granzymes A and B. Granzymes A and B were both shown to play an important role in LL-37-induced apoptosis of CTLs. Further analysis revealed that apoptosis occurred primarily through granzyme A-mediated caspase-independent apoptosis. However, caspase-dependent cell death was also observed. This suggests that LL-37 induces apoptosis in CTLs via multiple different mechanisms, initiated by the LL-37-induced leakage of granzymes from cytolytic granules. Our results imply the existence of a novel mechanism of crosstalk between the inflammatory and adaptive immune systems. Cells such as neutrophils, at the site of a tumor for example, could influence the effector, activity of CTL through the secretion of LL-37.
Collapse
|
17
|
Keech CL, Pang KC, McCluskey J, Chen W. Direct antigen presentation by DC shapes the functional CD8(+) T-cell repertoire against the nuclear self-antigen La-SSB. Eur J Immunol 2010; 40:330-8. [PMID: 19950171 DOI: 10.1002/eji.200939522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Controversy still surrounds the importance of cross-presentation versus endogenous or direct presentation of MHC-I restricted Ag in CD8(+) T-cell (T(CD8+)) immunity. It is even less clear what relative role these pathways play in shaping the T-cell repertoire specific for ubiquitous self-antigens, especially in cases where both Ag presentation pathways could potentially be involved. Here we provide evidence that a T(CD8+) repertoire specific for a determinant from the nuclear autoantigen La-SSB is largely shaped by direct presentation. In this system, mouse T(CD8+) reactive to a xenogeneic human La (hLa(51-58)) K(b) peptide did not recognize directly presented peptide on either spleen cells from hLa-Tg mice or hLa transfected syngeneic cells. Interestingly, the same T(CD8+) were activated by in vivo challenge with allogeneic APC expressing either the Tg hLa or loaded with intact recombinant hLa protein, indicating functional cross-presentation of the hLa(51-58). However, in irradiated bone marrow chimeric mice, DC expressing Tg hLa, but not WT DC that matured in hLa-Tg mice, constitutively presented the hLa(51-58) to T(CD8+). These data demonstrate that although both the direct- and cross-presentation pathways are potentially operative in revealing hLa(51-58) to T(CD8+), the T(CD8+) repertoire to this determinant is shaped quantitatively according to the efficiency of Ag presentation.
Collapse
Affiliation(s)
- Catherine L Keech
- Department of Microbiology and Immunology, University of Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
18
|
Austin ED, Rock MT, Mosse CA, Vnencak-Jones CL, Yoder SM, Robbins IM, Loyd JE, Meyrick BO. T lymphocyte subset abnormalities in the blood and lung in pulmonary arterial hypertension. Respir Med 2009; 104:454-62. [PMID: 19880300 DOI: 10.1016/j.rmed.2009.10.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Revised: 08/16/2009] [Accepted: 10/05/2009] [Indexed: 10/20/2022]
Abstract
RATIONALE Mounting data suggest that immune cell abnormalities participate in the pathogenesis of pulmonary arterial hypertension (PAH). OBJECTIVE To determine whether the T lymphocyte subset composition in the systemic circulation and peripheral lung is altered in PAH. METHODS Flow cytometric analyses were performed to determine the phenotypic profile of peripheral blood lymphocytes in idiopathic PAH (IPAH) patients (n=18) and healthy controls (n=17). Immunocytochemical analyses of lymphocytes and T cell subsets were used to examine lung tissue from PAH patients (n=11) and controls (n=11). MEASUREMENTS AND MAIN RESULTS IPAH patients have abnormal CD8+ T lymphocyte subsets, with a significant increase in CD45RA+ CCR7- peripheral cytotoxic effector-memory cells (p=0.02) and reduction of CD45RA+ CCR7+ naive CD8+ cells versus controls (p=0.001). Further, IPAH patients have a higher proportion of circulating regulatory T cells (T(reg)) and 4-fold increases in the number of CD3+ and CD8+ cells in the peripheral lung compared with controls (p<0.01). CONCLUSIONS Alterations in circulating T cell subsets, particularly CD8+ T lymphocytes and CD4+ T(reg), in patients with PAH suggest that a dysfunctional immune system contributes to disease pathogenesis. A preponderance of CD3+ and CD8+ T lymphocytes in the peripheral lung of PAH patients supports this concept.
Collapse
Affiliation(s)
- E D Austin
- Department of Pediatrics, Division of Pulmonary, Allergy, and Immunology Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Mortensen ES, Rekvig OP. Nephritogenic Potential of Anti-DNA Antibodies against Necrotic Nucleosomes. J Am Soc Nephrol 2009; 20:696-704. [DOI: 10.1681/asn.2008010112] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
20
|
Melanoma inhibitor of apoptosis protein (ML-IAP) specific cytotoxic T lymphocytes cross-react with an epitope from the auto-antigen SS56. J Invest Dermatol 2009; 129:1992-9. [PMID: 19212346 DOI: 10.1038/jid.2009.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A large proportion of melanoma patients host a spontaneous T-cell response specifically against ML-IAP-derived peptides. In this study, we describe that some ML-IAP-specific cytotoxic T cells isolated from melanoma patients cross react with an epitope from the auto-antigen SS56. SS56 is a recently described target of autoantibody responses in Sjögren's syndrome (SS) as well as systemic lupus erythematosus (SLE). Here, we describe that SS56 is also an auto-antigen for T cells in SS and SLE. Hence, SS56-specific T cells could readily be detected in circulation and among the infiltrating cells of SLE skin lesions. SS56-specific T cells were able to lyse target cells presenting the peptide epitope on the surface. Notably, SS56-specific CD8 T cells isolated from an SS patient cross reacted with the ML-IAP epitope. This early evidence of a target for auto-reactive CTL in SS and SLE patients; it is to our knowledge previously unreported and underscores the important role of CD8 T cells in autoimmune disorders. Furthermore, the cross-reactivity against the auto-antigen SS56 and the tumor-antigen ML-IAP confirms the link between autoimmunity and anti-cancer cellular immune responses.
Collapse
|
21
|
de Beukelaar JW, Sillevis Smitt PA, Hop WC, Kraan J, Hooijkaas H, Verjans GMGM, Gratama JW. Imbalances in circulating lymphocyte subsets in Hu antibody associated paraneoplastic neurological syndromes. Eur J Neurol 2008; 14:1383-91. [PMID: 18028190 DOI: 10.1111/j.1468-1331.2007.01986.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In paraneoplastic neurological syndromes (PNS) associated with small cell lung cancer (SCLC) and Hu antibodies, neuron-specific Hu antigens expressed by the tumour hypothetically trigger an immune response that cross-reacts with Hu antigens in the nervous system, resulting in tumour suppression and neuronal damage. To gain more insight into the hypothesized cell-mediated immune pathogenesis of these syndromes, we analysed the circulating lymphocyte subsets in untreated patients with SCLC, PNS and Hu antibodies (n = 18), SCLC without PNS (n = 19) and controls (n = 29) using flow cytometry. SCLC patients with PNS had a variety of imbalances within their circulating lymphocyte subsets as compared with SCLC patients without PNS and healthy controls: (i) a lymphopenia of the major subsets (i.e. B, CD4+ and CD8+ T lymphocytes); (ii) increased proportions of activated CD4+ and CD8+ T cells; (iii) reduced numbers of terminally differentiated effector CD8+ T cells and cells with a cytotoxic T-cell phenotype (CD56+ and CD57+). Although indirect, our data provide further support for the involvement of T cells in the pathogenesis of Hu antibody associated PNS.
Collapse
Affiliation(s)
- J W de Beukelaar
- Department of Medical Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
22
|
Liu G, Zhao Y. Toll-like receptors and immune regulation: their direct and indirect modulation on regulatory CD4+ CD25+ T cells. Immunology 2007; 122:149-56. [PMID: 17848162 PMCID: PMC2266004 DOI: 10.1111/j.1365-2567.2007.02651.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Regulatory CD4(+) CD25(+) T (Treg) cells with the ability to suppress host immune responses against self- or non-self antigens play important roles in the processes of autoimmunity, transplant rejection, infectious diseases and cancers. The proper regulation of CD4(+) CD25(+) Treg cells is thus critical for optimal immune responses. Toll-like receptor (TLR)-mediated recognition of specific structures of invading pathogens initiates innate as well as adaptive immune responses via antigen-presenting cells (APCs). Interestingly, new evidence suggests that TLR signalling may directly or indirectly regulate the immunosuppressive function of CD4(+) CD25(+) Treg cells in immune responses. TLR signalling may shift the balance between CD4(+) T-helper cells and Treg cells, and subsequently influence the outcome of the immune response. This immunomodulation pathway may therefore have potential applications in the treatment of graft rejection, autoimmune diseases, infection diseases and cancers.
Collapse
Affiliation(s)
- Guangwei Liu
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | |
Collapse
|
23
|
Mor A, Philips MR, Pillinger MH. The role of Ras signaling in lupus T lymphocytes: biology and pathogenesis. Clin Immunol 2007; 125:215-23. [PMID: 17913587 DOI: 10.1016/j.clim.2007.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 08/11/2007] [Accepted: 08/13/2007] [Indexed: 12/17/2022]
Abstract
Ras is a GTP-binding protein that plays multiple important roles in cell activation, including proliferative and inflammatory responses. Ras regulation is complex and depends upon post-translational processing, organelle-specific localization and the activation/deactivation of Ras by a number of regulatory molecules. Ras activation in T lymphocytes demonstrates unique features, including its dependence on the T cell receptor and the ability of Ras to signal from both the plasma membrane and the Golgi. Abnormalities of Ras expression, activation and signaling pathways in T lymphocytes appear to play important roles in the development of autoimmunity in general, and systemic lupus erythematosus in particular. In this manuscript, we review the basic biology of Ras in T lymphocytes, and the ways in which T lymphocyte Ras abnormalities may contribute to the development of a lupus phenotype.
Collapse
Affiliation(s)
- Adam Mor
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, the NYU Hospital for Joint Diseases, NY 10003, USA.
| | | | | |
Collapse
|
24
|
Bendiksen S, Mortensen ES, Olsen R, Fenton KA, Kalaaji M, Jørgensen L, Rekvig OP. Glomerular expression of large polyomavirus T antigen in binary tet-off regulated transgenic mice induces apoptosis, release of chromatin and initiates a lupus-like nephritis. Mol Immunol 2007; 45:728-39. [PMID: 17719634 DOI: 10.1016/j.molimm.2007.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2007] [Accepted: 07/03/2007] [Indexed: 11/20/2022]
Abstract
Binary tetracycline-regulated polyomavirus large T antigen transgenic mice were generated to study immunological tolerance for nucleosomes. Expression of T antigen resulted in binding of the protein to chromatin, and released T antigen-nucleosome complexes from dying cells maintained anti-dsDNA and anti-nucleosome antibody-production by activating autoimmune nucleosome-specific B cells and CD4+ and CD8+ T antigen specific T cells. Glomerular T antigen expression was observed in these mice. Here, we demonstrate that this expression was linked to glomerular cell apoptosis, release of nucleosomes and association of nucleosomes with glomerulus basement membranes, detected as electron dense structures. Immune electron microscopy (IEM) revealed that these structures were glomerular targets for induced anti-dsDNA and anti-T antigen antibodies. Co-localization IEM demonstrated that in vivo-bound auto-antibodies co-localized with experimental monoclonal antibodies to dsDNA and to T antigen. A comparative analysis of glomeruli from nephritic (NZWxNZB)F1 and T antigen expressing transgenic mice revealed deposition of nucleosomes in glomerular capillary and mesangial matrix membranes and binding of anti-nucleosome antibodies in both mice strains. A controlled experimental model that may elucidate the initial events accounting for nucleosome-mediated nephritis has not been available. The transgenic mouse may be important to describe early immunological and cellular events accounting for the enigmatic lupus nephritis.
Collapse
Affiliation(s)
- Signy Bendiksen
- Department of Medical Biochemistry, University Hospital of Northern Norway, N-9038 Tromsø, Norway
| | | | | | | | | | | | | |
Collapse
|
25
|
Blanco P, Pellegrin JL, Moreau JF, Viallard JF. Physiopathologie du lupus érythémateux systémique. Presse Med 2007; 36:825-34. [PMID: 17449371 DOI: 10.1016/j.lpm.2006.11.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2006] [Indexed: 01/08/2023] Open
Abstract
Innate immunity is directly implicated in the pathophysiology of lupus through the dendritic cell system and the activation by immune complexes of some toll-like receptors (TLR). Interferon-alpha plays a key role in the pathophysiology of lupus and represents a promising target for immune therapy. Dendritic cells are activated and able to capture large quantities of nuclear antigen-containing bodies to stimulate specific adaptive immune response.
Collapse
Affiliation(s)
- Patrick Blanco
- Clinique de médecine interne, Hôpital Haut-Lévêque, Pessac, France.
| | | | | | | |
Collapse
|
26
|
Getts MT, Kim BS, Miller SD. Differential outcome of tolerance induction in naive versus activated Theiler's virus epitope-specific CD8+ cytotoxic T cells. J Virol 2007; 81:6584-93. [PMID: 17428853 PMCID: PMC1900084 DOI: 10.1128/jvi.00008-07] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tolerance induced by the intravenous injection of peptide-pulsed, ethylene carbodiimide (ECDI)-fixed splenic antigen-presenting cells (Ag-SP) is a safe and effective method of inducing specific unresponsiveness in CD4+ T cells for the prevention and treatment of a variety of autoimmune diseases. We determined whether Ag-SP tolerance could also be used to tolerize CD8+ T cells. We show in the Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease model of multiple sclerosis that CD8+ T cells specific for both dominant and subdominant epitopes can be rendered tolerant. Interestingly, although virus clearance was delayed, lack of the virus-specific cytotoxic T-lymphocyte response did not result in the conversion of normally TMEV-resistant C57BL/6 mice to a susceptible phenotype. Importantly, we found that Ag-SP tolerance may not be a practical treatment for human diseases in which CD8+ T cells play a major role in pathogenesis, as tolerance induction in mice previously infected with TMEV led to a severe, often fatal reaction.
Collapse
Affiliation(s)
- Meghann Teague Getts
- Department of Microbiology and Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Avenue, Chicago, IL 60611, USA
| | | | | |
Collapse
|
27
|
Couzi L, Merville P, Deminière C, Moreau JF, Combe C, Pellegrin JL, Viallard JF, Blanco P. Predominance of CD8+ T lymphocytes among periglomerular infiltrating cells and link to the prognosis of class III and class IV lupus nephritis. ACTA ACUST UNITED AC 2007; 56:2362-70. [PMID: 17599764 DOI: 10.1002/art.22654] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Recent studies have revealed a potential implication of CD8+ T lymphocytes in the pathogenesis of systemic lupus erythematosus (SLE) through their ability to induce tissue damage. The aim of the present study was to analyze the localization of CD8+ cells in the kidneys of patients with class III and class IV lupus nephritis and to establish correlations with histologic, biologic, and clinical features of SLE. METHODS Twenty-five consecutive SLE patients with class III or class IV lupus nephritis were enrolled. Phenotype analyses of blood lymphocytes and renal immunohistochemistry studies were performed. RESULTS CD8+ T cells were the predominant kidney-infiltrating subset of cells. The mean +/- SD numbers of CD8+ T cells and CD4+ T cells were 66.2 +/- 65.2/mm(2) and 19.3 +/- 29.4/mm(2), respectively. There was a significant correlation between the percentage of blood CD3+,CD8+,DR+ cells and the total number of renal CD8+ T cells (r = 0.42, P = 0.039). Renal CD8+ T cell infiltration correlated well with the renal activity index (r = 0.63, P = 0.0007) and with high serum creatinine levels (r = 0.75, P = 0.0001). This CD8+ T cell infiltrate, which was predominantly in the periglomerular area, was correlated with cellular crescents and Bowman's capsule rupture and was associated with a poor response after conventional induction therapy. CONCLUSION CD8+ T lymphocytes infiltrate the periglomerular area in patients with severe (class III and class IV) lupus nephritis and are linked to a poor outcome after induction therapy. These results reveal a new potential effector pathway operant in lupus nephritis.
Collapse
Affiliation(s)
- Lionel Couzi
- CHU, and UMR-CNRS 5164, Université Victor Segalen Bordeaux 2, Bordeaux, France
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kurobe T, Hirono I, Kondo H, Saito-Taki T, Aoki T. Molecular cloning, characterization, expression and functional analysis of Japanese flounder Paralichthys olivaceus Fas ligand. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2007; 31:687-95. [PMID: 17197025 DOI: 10.1016/j.dci.2006.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Revised: 08/17/2006] [Accepted: 08/17/2006] [Indexed: 05/13/2023]
Abstract
We isolated and sequenced Fas ligand cDNA and its gene from Japanese flounder (JF), Paralichthys olivaceus. The JF-Fas ligand cDNA consisted of 1016 bp and encoded 230 amino acid residues. The identities of the deduced amino acid sequence of the JF-Fas ligand to human Fas ligand, Tumor necrosis factor-alpha and Lymphotoxin-alpha were 26.1%, 24.5% and 23.0%, respectively. A proline-rich domain (PRD) that is important for localization of the protein was found in the N-terminal region, and two cysteine residues, which form a disulfide bond, were conserved. The JF-Fas ligand gene has a length of 1.8 kb and consists of four exons and three introns. The length of the JF-Fas ligand second intron is shorter than that in the human and pig Fas ligand genes. However, the organization of the exons and introns is similar to that of mammals. RT-PCR was conducted for 12 tissues, and expression of JF-Fas ligand mRNA was detected in the kidney, thymus, gills, stomach and spleen. The recombinant JF-Fas ligand prepared in an Escherichia coli protein expression system showed cytotoxic activity against Japanese flounder cell line HINAE and caused the fragmentation of genomic DNA. The cytotoxic activity was measured by MTT assay. These results indicate that fish possess a Fas ligand system.
Collapse
Affiliation(s)
- Tomofumi Kurobe
- Laboratory of Genome Science, Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, Minato, Tokyo 108-8477, Japan
| | | | | | | | | |
Collapse
|
29
|
Kim R, Emi M, Tanabe K. Cancer immunosuppression and autoimmune disease: beyond immunosuppressive networks for tumour immunity. Immunology 2006; 119:254-64. [PMID: 17005005 PMCID: PMC1782355 DOI: 10.1111/j.1365-2567.2006.02430.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cancer immunosuppression evolves by constitution of an immunosuppressive network extending from a primary tumour site to secondary lymphoid organs and peripheral vessels and is mediated by several tumour-derived soluble factors (TDSFs) such as interleukin-10 (IL-10), transforming growth factor-beta (TGF-beta) and vascular endothelial growth factor (VEGF). TDSFs induce immature myeloid cells and regulatory T cells in accordance with tumour progression, resulting in the inhibition of dendritic cell maturation and T-cell activation in a tumour-specific immune response. Tumour cells grow by exploiting a pro-inflammatory situation in the tumour microenvironment, whereas immune cells are regulated by TDSFs during anti-inflammatory situations--mediated by impaired clearance of apoptotic cells--that cause the release of IL-10, TGF-beta, and prostaglandin E2 (PGE2) by macrophages. Accumulation of impaired apoptotic cells induces anti-DNA antibodies directed against self antigens, which resembles a pseudo-autoimmune status. Systemic lupus erythematosus is a prototype of autoimmune disease that is characterized by defective tolerance of self antigens, the presence of anti-DNA antibodies and a pro-inflammatory response. The anti-DNA antibodies can be produced by impaired clearance of apoptotic cells, which is the result of a hereditary deficiency of complements C1q, C3 and C4, which are involved in the recognition of phagocytosis by macrophages. Thus, it is likely that impaired clearance of apoptotic cells is able to provoke different types of immune dysfunction in cancer and autoimmune disease in which some are similar and others are critically different. This review discusses a comparison of immunological dysfunctions in cancer and autoimmune disease with the aim of exploring new insights beyond cancer immunosuppression in tumour immunity.
Collapse
Affiliation(s)
- Ryungsa Kim
- International Radiation Information Centre, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| | | | | |
Collapse
|
30
|
Kalaaji M, Mortensen E, Jørgensen L, Olsen R, Rekvig OP. Nephritogenic lupus antibodies recognize glomerular basement membrane-associated chromatin fragments released from apoptotic intraglomerular cells. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1779-92. [PMID: 16723695 PMCID: PMC1606630 DOI: 10.2353/ajpath.2006.051329] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Antibodies to dsDNA represent a classification criterion for systemic lupus erythematosus. Subpopulations of these antibodies are involved in lupus nephritis. No known marker separates nephritogenic from non-nephritogenic anti-dsDNA antibodies. It is not clear whether specificity for glomerular target antigens or intrinsic antibody-affinity for dsDNA or nucleosomes is a critical parameter. Furthermore, it is still controversial whether glomerular target antigen(s) is constituted by nucleosomes or by non-nucleosomal glomerular structures. Previously, we have demonstrated that antibodies eluted from murine nephritic kidneys recognize nucleosomes, but not other glomerular antigens. In this study, we determined the structures that bind nephritogenic autoantibodies in vivo by transmission electron microscopy, immune electron microscopy, and colocalization immune electron microscopy using experimental antibodies to dsDNA, to histones and transcription factors, or to laminin. The data obtained are consistent and point at glomerular basement membrane-associated nucleosomes as target structures for the nephritogenic autoantibodies. Terminal deoxynucleotidyl-transferase-mediated dUTP nick end-labeling or caspase-3 assays demonstrate that lupus nephritis is linked to intraglomerular cell apoptosis. The data suggest that nucleosomes are released by apoptosis and associate with glomerulus basement membranes, which may then be targeted by pathogenic anti-nucleosome antibodies. Thus, apoptotic nucleosomes may represent both inducer and target structures for nephritogenic autoantibodies in systemic lupus erythematosus.
Collapse
Affiliation(s)
- Manar Kalaaji
- Department of Biochemistry, Institute of Medical Biology, University of Tromsø, N-9037 Tromsø, Norway
| | | | | | | | | |
Collapse
|