1
|
Yandamuri SS, Filipek B, Lele N, Cohen I, Bennett JL, Nowak RJ, Sotirchos ES, Longbrake EE, Mace EM, O’Connor KC. A Noncanonical CD56dimCD16dim/- NK Cell Subset Indicative of Prior Cytotoxic Activity Is Elevated in Patients with Autoantibody-Mediated Neurologic Diseases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:785-800. [PMID: 38251887 PMCID: PMC10932911 DOI: 10.4049/jimmunol.2300015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD), myelin oligodendrocyte glycoprotein Ab disease, and autoimmune myasthenia gravis (MG) are autoantibody-mediated neurologic conditions where autoantibodies can induce Ab-dependent cellular cytotoxicity (ADCC), a NK cell-mediated effector function. However, whether ADCC is a pathogenic mechanism in patients with these conditions has not been confirmed. We sought to characterize circulatory NK cells using functional assays, phenotyping, and transcriptomics to elucidate their role in pathology. NK cells from NMOSD patients and MG patients with elevated disease burden exhibited reduced ADCC and CD56dimCD16hi NK cells, along with an elevated frequency of CD56dimCD16dim/- NK cells. We determined that ADCC induces a similar phenotypic shift in vitro. Bulk RNA sequencing distinguished the CD56dimCD16dim/- population from the canonical CD56dimCD16hi cytotoxic and CD56hiCD16- immunomodulatory subsets, as well as CD56hiCD16+ NK cells. Multiparameter immunophenotyping of NK cell markers, functional proteins, and receptors similarly showed that the CD56dimCD16dim/- subset exhibits a unique profile while still maintaining expression of characteristic NK markers CD56, CD94, and NKp44. Notably, expression of perforin and granzyme is reduced in comparison with CD56dimCD16hi NK cells. Moreover, they exhibit elevated trogocytosis capability, HLA-DR expression, and many chemokine receptors, including CCR7. In contrast with NMOSD and MG, myelin oligodendrocyte glycoprotein Ab disease NK cells did not exhibit functional, phenotypic, or transcriptomic perturbations. In summary, CD56dimCD16dim/- NK cells are a distinct peripheral blood immune cell population in humans elevated upon prior cytotoxic activity by the CD56dimCD16hi NK cell subset. The elevation of this subset in NMOSD and MG patients suggests prior ADCC activity.
Collapse
Affiliation(s)
- Soumya S. Yandamuri
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, United States
| | - Beata Filipek
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, United States
- Department of Pharmaceutical Microbiology and Biochemistry, Medical University of Lodz; Lodz, Poland
| | - Nikhil Lele
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
| | - Inessa Cohen
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
| | - Jeffrey L. Bennett
- Departments of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado School of Medicine, Anschutz Medical Campus; Aurora, CO, United States
| | - Richard J. Nowak
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
| | - Elias S. Sotirchos
- Department of Neurology, Johns Hopkins University; Baltimore, MD, United States
| | - Erin E. Longbrake
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
| | - Emily M. Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center; New York, NY, United States
| | - Kevin C. O’Connor
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, United States
| |
Collapse
|
2
|
Perri V, Zingaropoli MA, Pasculli P, Ciccone F, Tartaglia M, Baione V, Malimpensa L, Ferrazzano G, Mastroianni CM, Conte A, Ciardi MR. The Impact of Cytomegalovirus Infection on Natural Killer and CD8+ T Cell Phenotype in Multiple Sclerosis. BIOLOGY 2024; 13:154. [PMID: 38534424 DOI: 10.3390/biology13030154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/06/2024] [Accepted: 02/23/2024] [Indexed: 03/28/2024]
Abstract
Multiple sclerosis (MS) is a debilitating neurological disease that has been classified as an immune-mediated attack on myelin, the protective sheath of nerves. Some aspects of its pathogenesis are still unclear; nevertheless, it is generally established that viral infections influence the course of the disease. Cytomegalovirus (CMV) is a major pathogen involved in alterations of the immune system, including the expansion of highly differentiated cytotoxic CD8+ T cells and the accumulation of adaptive natural killer (NK) cells expressing high levels of the NKG2C receptor. In this study, we evaluated the impact of latent CMV infection on MS patients through the characterization of peripheral NK cells, CD8+ T cells, and NKT-like cells using flow cytometry. We evaluated the associations between immune cell profiles and clinical features such as MS duration and MS progression, evaluated using the Expanded Disability Status Scale (EDSS). We showed that NK cells, CD8+ T cells, and NKT-like cells had an altered phenotype in CMV-infected MS patients and displayed high levels of the NKG2C receptor. Moreover, in MS patients, increased NKG2C expression levels were found to be associated with higher EDSS scores. Overall, these results support the hypothesis that CMV infection imprints the immune system by modifying the phenotype and receptor repertoire of NK and CD8+ T cells, suggesting a detrimental role of CMV on MS progression.
Collapse
Affiliation(s)
- Valentina Perri
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | | | - Patrizia Pasculli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Federica Ciccone
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Matteo Tartaglia
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Viola Baione
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
| | | | - Gina Ferrazzano
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
| | | | - Antonella Conte
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
3
|
Hilliard KA, Throm AA, Pingel JT, Saucier N, Zaher HS, French AR. Expansion of a novel population of NK cells with low ribosome expression in juvenile dermatomyositis. Front Immunol 2022; 13:1007022. [PMID: 36389718 PMCID: PMC9660249 DOI: 10.3389/fimmu.2022.1007022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/10/2022] [Indexed: 02/06/2023] Open
Abstract
Juvenile dermatomyositis (JDM) is a pediatric autoimmune disease associated with characteristic rash and proximal muscle weakness. To gain insight into differential lymphocyte gene expression in JDM, peripheral blood mononuclear cells from 4 new-onset JDM patients and 4 healthy controls were sorted into highly enriched lymphocyte populations for RNAseq analysis. NK cells from JDM patients had substantially greater differentially expressed genes (273) than T (57) and B (33) cells. Upregulated genes were associated with the innate immune response and cell cycle, while downregulated genes were associated with decreased ribosomal RNA. Suppressed ribosomal RNA in JDM NK cells was validated by measuring transcription and phosphorylation levels. We confirmed a population of low ribosome expressing NK cells in healthy adults and children. This population of low ribosome NK cells was substantially expanded in 6 treatment-naïve JDM patients and was associated with decreased NK cell degranulation. The enrichment of this NK low ribosome population was completely abrogated in JDM patients with quiescent disease. Together, these data suggest NK cells are highly activated in new-onset JDM patients with an increased population of low ribosome expressing NK cells, which correlates with decreased NK cell function and resolved with control of active disease.
Collapse
Affiliation(s)
- Kinsey A. Hilliard
- Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Allison A. Throm
- Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
| | - Jeanette T. Pingel
- Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Nermina Saucier
- Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Hani S. Zaher
- Department of Biology, Washington University, St. Louis, MO, United States
| | - Anthony R. French
- Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
| |
Collapse
|
4
|
Wisgalla A, Ramien C, Streitz M, Schlickeiser S, Lupu AR, Diemert A, Tolosa E, Arck PC, Bellmann-Strobl J, Siebert N, Heesen C, Paul F, Friese MA, Infante-Duarte C, Gold SM. Alterations of NK Cell Phenotype During Pregnancy in Multiple Sclerosis. Front Immunol 2022; 13:907994. [PMID: 35860238 PMCID: PMC9289470 DOI: 10.3389/fimmu.2022.907994] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
In multiple sclerosis (MS), relapse rate is decreased by 70-80% in the third trimester of pregnancy. However, the underlying mechanisms driving this effect are poorly understood. Evidence suggests that CD56bright NK cell frequencies increase during pregnancy. Here, we analyze pregnancy-related NK cell shifts in a large longitudinal cohort of pregnant women with and without MS, and provide in-depth phenotyping of NK cells. In healthy pregnancy and pregnancy in MS, peripheral blood NK cells showed significant frequency shifts, notably an increase of CD56bright NK cells and a decrease of CD56dim NK cells toward the third trimester, indicating a general rather than an MS-specific phenomenon of pregnancy. Additional follow-ups in women with MS showed a reversal of NK cell changes postpartum. Moreover, high-dimensional profiling revealed a specific CD56bright subset with receptor expression related to cytotoxicity and cell activity (e.g., CD16+ NKp46high NKG2Dhigh NKG2Ahigh phenotype) that may drive the expansion of CD56bright NK cells during pregnancy in MS. Our data confirm that pregnancy promotes pronounced shifts of NK cells toward the regulatory CD56bright population. Although exploratory results on in-depth CD56bright phenotype need to be confirmed in larger studies, our findings suggest an increased regulatory NK activity, thereby potentially contributing to disease amelioration of MS during pregnancy.
Collapse
Affiliation(s)
- Anne Wisgalla
- Medizinische Klinik m.S. Psychosomatik, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Caren Ramien
- Institut für Neuroimmunologie und Multiple Sklerose (INIMS), Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Mathias Streitz
- Institut für Medizinische Immunologie, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Stephan Schlickeiser
- Institut für Medizinische Immunologie, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Andreea-Roxana Lupu
- Cantacuzino National Military Medical Institute for Research and Development, Bucharest, Romania
| | - Anke Diemert
- Klinik für Geburtshilfe und Pränatalmedizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Eva Tolosa
- Institut für Immunologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Petra C. Arck
- Klinik für Geburtshilfe und Pränatalmedizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Judith Bellmann-Strobl
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and Charité-Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Nadja Siebert
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Heesen
- Institut für Neuroimmunologie und Multiple Sklerose (INIMS), Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and Charité-Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Manuel A. Friese
- Institut für Neuroimmunologie und Multiple Sklerose (INIMS), Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Carmen Infante-Duarte
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan M. Gold
- Medizinische Klinik m.S. Psychosomatik, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- Institut für Neuroimmunologie und Multiple Sklerose (INIMS), Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
- Klinik für Psychiatrie und Psychotherapie, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- *Correspondence: Stefan M. Gold,
| |
Collapse
|
5
|
Beliën J, Goris A, Matthys P. Natural Killer Cells in Multiple Sclerosis: Entering the Stage. Front Immunol 2022; 13:869447. [PMID: 35464427 PMCID: PMC9019710 DOI: 10.3389/fimmu.2022.869447] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/14/2022] [Indexed: 11/14/2022] Open
Abstract
Studies investigating the immunopathology of multiple sclerosis (MS) have largely focused on adaptive T and B lymphocytes. However, in recent years there has been an increased interest in the contribution of innate immune cells, amongst which the natural killer (NK) cells. Apart from their canonical role of controlling viral infections, cell stress and malignancies, NK cells are increasingly being recognized for their modulating effect on the adaptive immune system, both in health and autoimmune disease. From different lines of research there is now evidence that NK cells contribute to MS immunopathology. In this review, we provide an overview of studies that have investigated the role of NK cells in the pathogenesis of MS by use of the experimental autoimmune encephalomyelitis (EAE) animal model, MS genetics or through ex vivo and in vitro work into the immunology of MS patients. With the advent of modern hypothesis-free technologies such as single-cell transcriptomics, we are exposing an unexpected NK cell heterogeneity, increasingly blurring the boundaries between adaptive and innate immunity. We conclude that unravelling this heterogeneity, as well as the mechanistic link between innate and adaptive immune cell functions will lay the foundation for the use of NK cells as prognostic tools and therapeutic targets in MS and a myriad of other currently uncurable autoimmune disorders.
Collapse
Affiliation(s)
- Jarne Beliën
- Department of Neurosciences, Laboratory for Neuroimmunology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - An Goris
- Department of Neurosciences, Laboratory for Neuroimmunology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Calahorra L, Camacho-Toledano C, Serrano-Regal MP, Ortega MC, Clemente D. Regulatory Cells in Multiple Sclerosis: From Blood to Brain. Biomedicines 2022; 10:335. [PMID: 35203544 PMCID: PMC8961785 DOI: 10.3390/biomedicines10020335] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune, and neurodegenerative disease of the central nervous system (CNS) that affects myelin. The etiology of MS is unclear, although a variety of environmental and genetic factors are thought to increase the risk of developing the disease. Historically, T cells were considered to be the orchestrators of MS pathogenesis, but evidence has since accumulated implicating B lymphocytes and innate immune cells in the inflammation, demyelination, and axonal damage associated with MS disease progression. However, more recently the importance of the protective role of immunoregulatory cells in MS has become increasingly evident, such as that of myeloid-derived suppressor cells (MDSCs), regulatory T (Treg) and B (Breg) cells, or CD56bright natural killer cells. In this review, we will focus on how peripheral regulatory cells implicated in innate and adaptive immune responses are involved in the physiopathology of MS. Moreover, we will discuss how these cells are thought to act and contribute to MS histopathology, also addressing their promising role as promoters of successful remyelination within the CNS. Finally, we will analyze how understanding these protective mechanisms may be crucial in the search for potential therapies for MS.
Collapse
Affiliation(s)
| | | | | | | | - Diego Clemente
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071 Toledo, Spain; (L.C.); (C.C.-T.); (M.P.S.-R.); (M.C.O.)
| |
Collapse
|
7
|
Capturing SNP Association across the NK Receptor and HLA Gene Regions in Multiple Sclerosis by Targeted Penalised Regression Models. Genes (Basel) 2021; 13:genes13010087. [PMID: 35052430 PMCID: PMC8774935 DOI: 10.3390/genes13010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 11/17/2022] Open
Abstract
Conventional genome-wide association studies (GWASs) of complex traits, such as Multiple Sclerosis (MS), are reliant on per-SNP p-values and are therefore heavily burdened by multiple testing correction. Thus, in order to detect more subtle alterations, ever increasing sample sizes are required, while ignoring potentially valuable information that is readily available in existing datasets. To overcome this, we used penalised regression incorporating elastic net with a stability selection method by iterative subsampling to detect the potential interaction of loci with MS risk. Through re-analysis of the ANZgene dataset (1617 cases and 1988 controls) and an IMSGC dataset as a replication cohort (1313 cases and 1458 controls), we identified new association signals for MS predisposition, including SNPs above and below conventional significance thresholds while targeting two natural killer receptor loci and the well-established HLA loci. For example, rs2844482 (98.1% iterations), otherwise ignored by conventional statistics (p = 0.673) in the same dataset, was independently strongly associated with MS in another GWAS that required more than 40 times the number of cases (~45 K). Further comparison of our hits to those present in a large-scale meta-analysis, confirmed that the majority of SNPs identified by the elastic net model reached conventional statistical GWAS thresholds (p < 5 × 10−8) in this much larger dataset. Moreover, we found that gene variants involved in oxidative stress, in addition to innate immunity, were associated with MS. Overall, this study highlights the benefit of using more advanced statistical methods to (re-)analyse subtle genetic variation among loci that have a biological basis for their contribution to disease risk.
Collapse
|
8
|
Fernandes SB, Patil ND, Meriaux S, Theresine M, Muller CP, Leenen FAD, Elwenspoek MMC, Zimmer J, Turner JD. Unbiased Screening Identifies Functional Differences in NK Cells After Early Life Psychosocial Stress. Front Immunol 2021; 12:674532. [PMID: 34394074 PMCID: PMC8363253 DOI: 10.3389/fimmu.2021.674532] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022] Open
Abstract
Early Life Adversity (ELA) is closely associated with the risk for developing diseases later in life, such as autoimmune diseases, type-2 diabetes and cardiovascular diseases. In humans, early parental separation, physical and sexual abuse or low social-economic status during childhood are known to have great impact on brain development, in the hormonal system and immune responses. Maternal deprivation (MD) is the closest animal model available to the human situation. This paradigm induces long lasting behavioral effects, causes changes in the HPA axis and affects the immune system. However, the mechanisms underlying changes in the immune response after ELA are still not fully understood. In this study we investigated how ELA changes the immune system, through an unbiased analysis, viSNE, and addressed specially the NK immune cell population and its functionality. We have demonstrated that maternal separation, in both humans and rats, significantly affects the sensitivity of the immune system in adulthood. Particularly, NK cells’ profile and response to target cell lines are significantly changed after ELA. These immune cells in rats are not only less cytotoxic towards YAC-1 cells, but also show a clear increase in the expression of maturation markers after 3h of maternal separation. Similarly, individuals who suffered from ELA display significant changes in the cytotoxic profile of NK cells together with decreased degranulation capacity. These results suggest that one of the key mechanisms by which the immune system becomes impaired after ELA might be due to a shift on the senescent state of the cells, specifically NK cells. Elucidation of such a mechanism highlights the importance of ELA prevention and how NK targeted immunotherapy might help attenuating ELA consequences.
Collapse
Affiliation(s)
- Sara B Fernandes
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Doctoral School in Systems and Molecular Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Neha D Patil
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Doctoral School in Systems and Molecular Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sophie Meriaux
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Maud Theresine
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Claude P Muller
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Fleur A D Leenen
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Martha M C Elwenspoek
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Jacques Zimmer
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Doctoral School in Systems and Molecular Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jonathan D Turner
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
9
|
Schwichtenberg SC, Wisgalla A, Schroeder-Castagno M, Alvarez-González C, Schlickeiser S, Siebert N, Bellmann-Strobl J, Wernecke KD, Paul F, Dörr J, Infante-Duarte C. Fingolimod Therapy in Multiple Sclerosis Leads to the Enrichment of a Subpopulation of Aged NK Cells. Neurotherapeutics 2021; 18:1783-1797. [PMID: 34244929 PMCID: PMC8608997 DOI: 10.1007/s13311-021-01078-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 02/04/2023] Open
Abstract
Fingolimod is an approved oral treatment for relapsing-remitting multiple sclerosis (RRMS) that modulates agonistically the sphingosin-1-phosphate receptor (S1PR), inhibiting thereby the egress of lymphocytes from the lymph nodes. In this interventional prospective clinical phase IV trial, we longitudinally investigated the impact of fingolimod on frequencies of NK cell subpopulations by flow cytometry in 17 RRMS patients at baseline and 1, 3, 6, and 12 months after treatment initiation. Clinical outcome was assessed by the Expanded Disability Status Scale (EDSS) and annualized relapse rates (ARR). Over the study period, median EDSS remained stable from month 3 to month 12, and ARR decreased compared to ARR in the 24 months prior treatment. Treatment was paralleled by an increased frequency of circulating NK cells, due primarily to an increase in CD56dimCD94low mature NK cells, while the CD56bright fraction and CD127+ innate lymphoid cells (ILCs) decreased over time. An unsupervised clustering algorithm further revealed that a particular fraction of NK cells defined by the expression of CD56dimCD16++KIR+/-NKG2A-CD94-CCR7+/-CX3CR1+/-NKG2C-NKG2D+NKp46-DNAM1++CD127+ increased during treatment. This specific phenotype might reflect a status of aged, fully differentiated, and less functional NK cells. Our study confirms that fingolimod treatment affects both NK cells and ILC. In addition, our study suggests that treatment leads to the enrichment of a specific NK cell subset characterized by an aged phenotype. This might limit the anti-microbial and anti-tumour NK cell activity in fingolimod-treated patients.
Collapse
Affiliation(s)
- Svenja C Schwichtenberg
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Augustenburger Platz 1 (Südstr. 2/Föhrer Str. 15), 13353, Berlin, Germany
| | - Anne Wisgalla
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Augustenburger Platz 1 (Südstr. 2/Föhrer Str. 15), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for "Psychiatrie Und Medizinische Klinik M.S. Psychosomatik,", Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Maria Schroeder-Castagno
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Augustenburger Platz 1 (Südstr. 2/Föhrer Str. 15), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
| | - Cesar Alvarez-González
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Augustenburger Platz 1 (Südstr. 2/Föhrer Str. 15), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
| | - Stephan Schlickeiser
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Föhrer Str. 15, 13353, Berlin, Germany
| | - Nadja Siebert
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine & Charité - Universitätsmedizin Berlin, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Judith Bellmann-Strobl
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine & Charité - Universitätsmedizin Berlin, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Klaus-Dieter Wernecke
- Charité - Universitätsmedizin Berlin and CRO SOSTANA GmbH, Wildensteiner Straße 27, 10318, Berlin, Germany
| | - Friedemann Paul
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine & Charité - Universitätsmedizin Berlin, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Jan Dörr
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine & Charité - Universitätsmedizin Berlin, Robert-Rössle-Straße 10, 13125, Berlin, Germany
- Current Affiliation: Multiple Sclerosis Center, Oberhavel Kliniken, Marwitzer Straße 91, 16761, Hennigsdorf, Germany
| | - Carmen Infante-Duarte
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Augustenburger Platz 1 (Südstr. 2/Föhrer Str. 15), 13353, Berlin, Germany.
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine & Charité - Universitätsmedizin Berlin, Robert-Rössle-Straße 10, 13125, Berlin, Germany.
| |
Collapse
|
10
|
Kucuksezer UC, Aktas Cetin E, Esen F, Tahrali I, Akdeniz N, Gelmez MY, Deniz G. The Role of Natural Killer Cells in Autoimmune Diseases. Front Immunol 2021; 12:622306. [PMID: 33717125 PMCID: PMC7947192 DOI: 10.3389/fimmu.2021.622306] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Natural killer (NK) cells, the large granular lymphocytes differentiated from the common lymphoid progenitors, were discovered in early 1970's. They are members of innate immunity and were initially defined by their strong cytotoxicity against virus-infected cells and by their important effector functions in anti-tumoral immune responses. Nowadays, NK cells are classified among the recently discovered innate lymphoid cell subsets and have capacity to influence both innate and adaptive immune responses. Therefore, they can be considered as innate immune cells that stands between the innate and adaptive arms of immunity. NK cells don't express T or B cell receptors and are recognized by absence of CD3. There are two major subgroups of NK cells according to their differential expression of CD16 and CD56. While CD16+CD56dim subset is best-known by their cytotoxic functions, CD16-CD56bright NK cell subset produces a bunch of cytokines comparable to CD4+ T helper cell subsets. Another subset of NK cells with production of interleukin (IL)-10 was named as NK regulatory cells, which has suppressive properties and could take part in immune-regulatory responses. Activation of NK cells is determined by a delicate balance of cell-surface receptors that have either activating or inhibitory properties. On the other hand, a variety of cytokines including IL-2, IL-12, IL-15, and IL-18 influence NK cell activity. NK-derived cytokines and their cytotoxic functions through induction of apoptosis take part in regulation of the immune responses and could contribute to the pathogenesis of many immune mediated diseases including ankylosing spondylitis, Behçet's disease, multiple sclerosis, rheumatoid arthritis, psoriasis, systemic lupus erythematosus and type-1 diabetes. Dysregulation of NK cells in autoimmune disorders may occur through multiple mechanisms. Thanks to the rapid developments in biotechnology, progressive research in immunology enables better characterization of cells and their delicate roles in the complex network of immunity. As NK cells stand in between innate and adaptive arms of immunity and "bridge" them, their contribution in inflammation and immune regulation deserves intense investigations. Better understanding of NK-cell biology and their contribution in both exacerbation and regulation of inflammatory disorders is a requisite for possible utilization of these multi-faceted cells in novel therapeutic interventions.
Collapse
Affiliation(s)
- Umut Can Kucuksezer
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Esin Aktas Cetin
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Fehim Esen
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
- Department of Ophthalmology, Medical Faculty, Istanbul Medeniyet University, Istanbul, Turkey
| | - Ilhan Tahrali
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Nilgun Akdeniz
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Metin Yusuf Gelmez
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Gunnur Deniz
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
11
|
Yang Y, Day J, Souza-Fonseca Guimaraes F, Wicks IP, Louis C. Natural killer cells in inflammatory autoimmune diseases. Clin Transl Immunology 2021; 10:e1250. [PMID: 33552511 PMCID: PMC7850912 DOI: 10.1002/cti2.1250] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells are a specialised population of innate lymphoid cells (ILCs) that help control local immune responses. Through natural cytotoxicity, production of cytokines and chemokines, and migratory capacity, NK cells play a vital immunoregulatory role in the initiation and chronicity of inflammatory and autoimmune responses. Our understanding of their functional differences and contributions in disease settings is evolving owing to new genetic and functional murine proof-of-concept studies. Here, we summarise current understanding of NK cells in several classic autoimmune disorders, particularly in rheumatoid arthritis (RA), multiple sclerosis (MS), systemic lupus erythematosus (SLE) and type 1 diabetes mellitus (T1DM), but also less understood diseases such as idiopathic inflammatory myopathies (IIMs). A better understanding of how NK cells contribute to these autoimmune disorders may pave the way for NK cell-targeted therapeutics.
Collapse
Affiliation(s)
- Yuyan Yang
- Tsinghua University School of Medicine Beijing China.,Inflammation Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | - Jessica Day
- Inflammation Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia.,Medical Biology University of Melbourne Melbourne VIC Australia.,Rheumatology Unit The Royal Melbourne Hospital Parkville VIC Australia
| | | | - Ian P Wicks
- Inflammation Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia.,Medical Biology University of Melbourne Melbourne VIC Australia.,Rheumatology Unit The Royal Melbourne Hospital Parkville VIC Australia
| | - Cynthia Louis
- Inflammation Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia.,Medical Biology University of Melbourne Melbourne VIC Australia
| |
Collapse
|
12
|
A CD8 + NK cell transcriptomic signature associated with clinical outcome in relapsing remitting multiple sclerosis. Nat Commun 2021; 12:635. [PMID: 33504809 PMCID: PMC7840761 DOI: 10.1038/s41467-020-20594-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) with the majority of cases characterised by relapsing/remitting (RRMS) attacks of neurologic dysfunction followed by variable resolution. Improving clinical outcomes in RRMS requires both a better understanding of the immunological mechanisms driving recurrent demyelination and better means of predicting future disease course to facilitate early targeted therapy. Here, we apply hypothesis-generating network transcriptomics to CD8+ cells isolated from patients in RRMS, identifying a signature reflecting expansion of a subset of CD8+ natural killer cells (NK8+) associated with favourable outcome. NK8+ are capable of regulating CD4+ T cell activation and proliferation in vitro, with reduced expression of HLA-G binding inhibitory receptors and consequent reduced sensitivity to HLA-G-mediated suppression. We identify surrogate markers of the NK8+ signature in peripheral blood leucocytes and validate their association with clinical outcome in an independent cohort, suggesting their measurement may facilitate early, targeted therapy in RRMS.
Collapse
|
13
|
van Eeden C, Khan L, Osman MS, Cohen Tervaert JW. Natural Killer Cell Dysfunction and Its Role in COVID-19. Int J Mol Sci 2020; 21:E6351. [PMID: 32883007 PMCID: PMC7503862 DOI: 10.3390/ijms21176351] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
When facing an acute viral infection, our immune systems need to function with finite precision to enable the elimination of the pathogen, whilst protecting our bodies from immune-related damage. In many instances however this "perfect balance" is not achieved, factors such as ageing, cancer, autoimmunity and cardiovascular disease all skew the immune response which is then further distorted by viral infection. In SARS-CoV-2, although the vast majority of COVID-19 cases are mild, as of 24 August 2020, over 800,000 people have died, many from the severe inflammatory cytokine release resulting in extreme clinical manifestations such as acute respiratory distress syndrome (ARDS) and hemophagocytic lymphohistiocytosis (HLH). Severe complications are more common in elderly patients and patients with cardiovascular diseases. Natural killer (NK) cells play a critical role in modulating the immune response and in both of these patient groups, NK cell effector functions are blunted. Preliminary studies in COVID-19 patients with severe disease suggests a reduction in NK cell number and function, resulting in decreased clearance of infected and activated cells, and unchecked elevation of tissue-damaging inflammation markers. SARS-CoV-2 infection skews the immune response towards an overwhelmingly inflammatory phenotype. Restoration of NK cell effector functions has the potential to correct the delicate immune balance required to effectively overcome SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | | | | | - Jan Willem Cohen Tervaert
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (C.v.E.); (L.K.); (M.S.O.)
| |
Collapse
|
14
|
Anderson KM, Augusto DG, Dandekar R, Shams H, Zhao C, Yusufali T, Montero-Martín G, Marin WM, Nemat-Gorgani N, Creary LE, Caillier S, Mofrad MRK, Parham P, Fernández-Viña M, Oksenberg JR, Norman PJ, Hollenbach JA. Killer Cell Immunoglobulin-like Receptor Variants Are Associated with Protection from Symptoms Associated with More Severe Course in Parkinson Disease. THE JOURNAL OF IMMUNOLOGY 2020; 205:1323-1330. [PMID: 32709660 DOI: 10.4049/jimmunol.2000144] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/28/2020] [Indexed: 12/12/2022]
Abstract
Immune dysfunction plays a role in the development of Parkinson disease (PD). NK cells regulate immune functions and are modulated by killer cell immunoglobulin-like receptors (KIR). KIR are expressed on the surface of NK cells and interact with HLA class I ligands on the surface of all nucleated cells. We investigated KIR-allelic polymorphism to interrogate the role of NK cells in PD. We sequenced KIR genes from 1314 PD patients and 1978 controls using next-generation methods and identified KIR genotypes using custom bioinformatics. We examined associations of KIR with PD susceptibility and disease features, including age at disease onset and clinical symptoms. We identified two KIR3DL1 alleles encoding highly expressed inhibitory receptors associated with protection from PD clinical features in the presence of their cognate ligand: KIR3DL1*015/HLA-Bw4 from rigidity (p c = 0.02, odds ratio [OR] = 0.39, 95% confidence interval [CI] 0.23-0.69) and KIR3DL1*002/HLA-Bw4i from gait difficulties (p c = 0.05, OR = 0.62, 95% CI 0.44-0.88), as well as composite symptoms associated with more severe disease. We also developed a KIR3DL1/HLA interaction strength metric and found that weak KIR3DL1/HLA interactions were associated with rigidity (pc = 0.05, OR = 9.73, 95% CI 2.13-172.5). Highly expressed KIR3DL1 variants protect against more debilitating symptoms of PD, strongly implying a role of NK cells in PD progression and manifestation.
Collapse
Affiliation(s)
- Kirsten M Anderson
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158
| | - Danillo G Augusto
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158
| | - Ravi Dandekar
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158
| | - Hengameh Shams
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158
| | - Chao Zhao
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158
| | - Tasneem Yusufali
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158
| | | | - Wesley M Marin
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158
| | - Neda Nemat-Gorgani
- Department of Structural Biology and Immunology, Stanford University, Palo Alto, CA 94305
| | - Lisa E Creary
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA 94304
| | - Stacy Caillier
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Department of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA 94720; and
| | - Peter Parham
- Department of Structural Biology and Immunology, Stanford University, Palo Alto, CA 94305
| | | | - Jorge R Oksenberg
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158
| | - Paul J Norman
- Division of Biomedical Informatics and Personalized Medicine, Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Jill A Hollenbach
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158;
| |
Collapse
|
15
|
McQuaid SL, Loughran ST, Power PA, Maguire P, Szczygiel A, Johnson PA. Low-dose IL-2 induces CD56 bright NK regulation of T cells via NKp44 and NKp46. Clin Exp Immunol 2020; 200:228-241. [PMID: 31989589 PMCID: PMC7232012 DOI: 10.1111/cei.13422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2020] [Indexed: 12/16/2022] Open
Abstract
Low-dose interleukin (IL)-2 has shown clinical benefits in patients with autoimmune and inflammatory diseases. Both regulatory T cells (Tregs ) and natural killer (NK) cells are increased in response to low-dose IL-2 immunotherapy. The role of regulatory T cells in autoimmune diseases has been extensively studied; however, NK cells have not been as thoroughly explored. It has not been well reported whether the increase in NK cells is purely an epiphenomenon or carries actual benefits for patients with autoimmune diseases. We demonstrate that low-dose IL-2 expands the primary human CD56bright NK cells resulting in a contact-dependent cell cycle arrest of effector T cells (Teffs ) via retention of the cycle inhibitor p21. We further show that NK cells respond via IL-2R-β, which has been shown to be significant for immunity by regulating T cell expansion. Moreover, we demonstrate that blocking NK receptors NKp44 and NKp46 but not NKp30 could abrogate the regulation of proliferation associated with low-dose IL-2. The increase in NK cells was also accompanied by an increase in Treg cells, which is dependent on the presence of CD56bright NK cells. These results not only heighten the importance of NK cells in low-dose IL-2 therapy but also identify key human NK targets, which may provide further insights into the therapeutic mechanisms of low-dose IL-2 in autoimmunity.
Collapse
Affiliation(s)
- S. L. McQuaid
- Viral Immunology LaboratorySchool of Nursing, Psychotherapy and Community HealthDublin City UniversityDublinIreland
- Mason Technology LtdDublinIreland
| | - S. T. Loughran
- Viral Immunology LaboratorySchool of Nursing, Psychotherapy and Community HealthDublin City UniversityDublinIreland
- Department of Applied ScienceDundalk Institute of TechnologyDundalkIreland
| | - P. A. Power
- Viral Immunology LaboratorySchool of Nursing, Psychotherapy and Community HealthDublin City UniversityDublinIreland
- Technological University DublinDublinIreland
| | - P. Maguire
- Viral Immunology LaboratorySchool of Nursing, Psychotherapy and Community HealthDublin City UniversityDublinIreland
- School of BiotechnologyDublin City UniversityDublinIreland
| | - A. Szczygiel
- Viral Immunology LaboratorySchool of Nursing, Psychotherapy and Community HealthDublin City UniversityDublinIreland
| | - P. A. Johnson
- Viral Immunology LaboratorySchool of Nursing, Psychotherapy and Community HealthDublin City UniversityDublinIreland
| |
Collapse
|
16
|
Banerjee PP, Pang L, Soldan SS, Miah SM, Eisenberg A, Maru S, Waldman A, Smith EA, Rosenberg-Hasson Y, Hirschberg D, Smith A, Ablashi DV, Campbell KS, Orange JS. KIR2DL4-HLAG interaction at human NK cell-oligodendrocyte interfaces regulates IFN-γ-mediated effects. Mol Immunol 2019; 115:39-55. [PMID: 30482463 PMCID: PMC6543535 DOI: 10.1016/j.molimm.2018.09.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 09/11/2018] [Accepted: 09/30/2018] [Indexed: 12/12/2022]
Abstract
Interactions between germline-encoded natural killer (NK) cell receptors and their respective ligands on tumorigenic or virus-infected cells determine NK cell cytotoxic activity and/or cytokine secretion. NK cell cytokine responses can be augmented in and can potentially contribute to multiple sclerosis (MS), an inflammatory disease of the central nervous system focused upon the oligodendrocytes (OLs). To investigate mechanisms by which NK cells may contribute to MS pathogenesis, we developed an in vitro human model of OL-NK cell interaction. We found that activated, but not resting human NK cells form conjugates with, and mediate cytotoxicity against, human oligodendrocytes. NK cells, when in conjugate with OLs, rapidly synthesize and polarize IFN-γ toward the OLs. IFN-γ is capable of reducing myelin oligodendrocyte and myelin associated glycoproteins (MOG and MAG) content. This activity is independent of MHC class-I mediated inhibition via KIR2DL1, but dependent upon the interaction between NK cell-expressed KIR2DL4 and its oligodendrocyte-expressed ligand, HLA-G. NK cells from patients with MS express higher levels of IFN-γ following conjugation to OLs, more actively promote in vitro reduction of MOG and MAG and have higher frequencies of the KIR2DL4 positive population. These data collectively suggest a mechanism by which NK cells can promote pathogenic effects upon OLs.
Collapse
Affiliation(s)
- P P Banerjee
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA.
| | - L Pang
- Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| | - S S Soldan
- The Wistar Institute, 3601 Spruce St., Philadelphia, PA 19104, USA
| | - S M Miah
- Blood Cell Development and Function Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - A Eisenberg
- The Children's Hospital of Philadelphia Research Institute, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - S Maru
- The Children's Hospital of Philadelphia Research Institute, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - A Waldman
- The Children's Hospital of Philadelphia Research Institute, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - E A Smith
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| | - Y Rosenberg-Hasson
- Human Immune Monitoring Center, Stanford School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA
| | - D Hirschberg
- Human Immune Monitoring Center, Stanford School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA
| | - A Smith
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| | - D V Ablashi
- Human Herpes Virus 6 Foundation, 1482 East Valley Road, Suite 619 Santa Barbara, CA 93108, USA
| | - K S Campbell
- Blood Cell Development and Function Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - J S Orange
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| |
Collapse
|
17
|
Wang XS, Cao F, Zhang Y, Pan HF. Therapeutic potential of aryl hydrocarbon receptor in autoimmunity. Inflammopharmacology 2019; 28:63-81. [PMID: 31617124 DOI: 10.1007/s10787-019-00651-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/30/2019] [Indexed: 12/11/2022]
Abstract
Aryl hydrocarbon receptor (AhR), a type of transcriptional factor, is widely expressed in immune cells. The activation of AhR signaling pathway depends on its ligands, which exist in environment and can also be produced by metabolism. Normal expressions of AhR and AhR-mediated signaling may be essential for immune responses, and effects of AhR signaling on the development and function of innate and adaptive immune cells have also been revealed in previous studies. Recent studies also indicate that aberrant AhR signaling may be related to autoimmune diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), multiple sclerosis (MS), autoimmune uveitis (AU), autoimmune diabetes, Behcet's disease (BD) and myasthenia gravis (MG). Moreover, administration of AhR ligands or drugs has been proven effective for improving pathological outcomes in some autoimmune diseases or models. In this review, we summarize the effects of AhR on several innate and adaptive immune cells associated with autoimmunity, and the mechanism on how AhR participates in autoimmune diseases. In addition, we also discuss therapeutic potential and application prospect of AhR in autoimmune diseases, so as to provide valuable information for exploring novel and effective approaches to autoimmune disease treatments.
Collapse
Affiliation(s)
- Xiao-Song Wang
- The First Affiliated Hospital of Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.,Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China
| | - Fan Cao
- Department of Clinical Medicine, The Second School of Clinical Medicine, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Yi Zhang
- Reproductive Medicine Center, Anhui Women and Child Health Care Hospital, 15 Yimin Street, Hefei, Anhui, 230011, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China. .,Anhui Province Key Laboratory of Major Autoimmune Diseases, 81 Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
18
|
Ott M, Avendaño-Guzmán E, Ullrich E, Dreyer C, Strauss J, Harden M, Schön M, Schön MP, Bernhardt G, Stadelmann C, Wegner C, Brück W, Nessler S. Laquinimod, a prototypic quinoline-3-carboxamide and aryl hydrocarbon receptor agonist, utilizes a CD155-mediated natural killer/dendritic cell interaction to suppress CNS autoimmunity. J Neuroinflammation 2019; 16:49. [PMID: 30808363 PMCID: PMC6390632 DOI: 10.1186/s12974-019-1437-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/17/2019] [Indexed: 12/18/2022] Open
Abstract
Background Quinoline-3-carboxamides, such as laquinimod, ameliorate CNS autoimmunity in patients and reduce tumor cell metastasis experimentally. Previous studies have focused on the immunomodulatory effect of laquinimod on myeloid cells. The data contained herein suggest that quinoline-3-carboxamides improve the immunomodulatory and anti-tumor effects of NK cells by upregulating the adhesion molecule DNAX accessory molecule-1 (DNAM-1). Methods We explored how NK cell activation by laquinimod inhibits CNS autoimmunity in experimental autoimmune encephalomyelitis (EAE), the most utilized model of MS, and improves immunosurveillance of experimental lung melanoma metastasis. Functional manipulations included in vivo NK and DC depletion experiments and in vitro assays of NK cell function. Clinical, histological, and flow cytometric read-outs were assessed. Results We demonstrate that laquinimod activates natural killer (NK) cells via the aryl hydrocarbon receptor and increases their DNAM-1 cell surface expression. This activation improves the cytotoxicity of NK cells against B16F10 melanoma cells and augments their immunoregulatory functions in EAE by interacting with CD155+ dendritic cells (DC). Noteworthy, the immunosuppressive effect of laquinimod-activated NK cells was due to decreasing MHC class II antigen presentation by DC and not by increasing DC killing. Conclusions This study clarifies how DNAM-1 modifies the bidirectional crosstalk of NK cells with CD155+ DC, which can be exploited to suppress CNS autoimmunity and strengthen tumor surveillance. Electronic supplementary material The online version of this article (10.1186/s12974-019-1437-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Martina Ott
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Erika Avendaño-Guzmán
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Evelyn Ullrich
- LOEWE Center for Cell and Gene Therapy, Goethe University, Frankfurt am Main, Germany.,Division of Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine, Hospital of the Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Carolin Dreyer
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Judith Strauss
- Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, Göttingen, Germany
| | - Markus Harden
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Margarete Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany.,Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen and University of Osnabrück, Göttingen, Germany
| | - Günter Bernhardt
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Gebäude I11 OE 5240, 30625, Hannover, Germany
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Christiane Wegner
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Present Address: Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Nessler
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
19
|
Presta I, Vismara M, Novellino F, Donato A, Zaffino P, Scali E, Pirrone KC, Spadea MF, Malara N, Donato G. Innate Immunity Cells and the Neurovascular Unit. Int J Mol Sci 2018; 19:E3856. [PMID: 30513991 PMCID: PMC6321635 DOI: 10.3390/ijms19123856] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
Recent studies have clarified many still unknown aspects related to innate immunity and the blood-brain barrier relationship. They have also confirmed the close links between effector immune system cells, such as granulocytes, macrophages, microglia, natural killer cells and mast cells, and barrier functionality. The latter, in turn, is able to influence not only the entry of the cells of the immune system into the nervous tissue, but also their own activation. Interestingly, these two components and their interactions play a role of great importance not only in infectious diseases, but in almost all the pathologies of the central nervous system. In this paper, we review the main aspects in the field of vascular diseases (cerebral ischemia), of primitive and secondary neoplasms of Central Nervous System CNS, of CNS infectious diseases, of most common neurodegenerative diseases, in epilepsy and in demyelinating diseases (multiple sclerosis). Neuroinflammation phenomena are constantly present in all diseases; in every different pathological state, a variety of innate immunity cells responds to specific stimuli, differentiating their action, which can influence the blood-brain barrier permeability. This, in turn, undergoes anatomical and functional modifications, allowing the stabilization or the progression of the pathological processes.
Collapse
Affiliation(s)
- Ivan Presta
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Marco Vismara
- Department of Cell Biotechnologies and Hematology, University "La Sapienza" of Rome, 00185 Rome, Italy.
| | - Fabiana Novellino
- Institute of Molecular Bioimaging and Physiology, National Research Council, 88100 Catanzaro, Italy.
| | - Annalidia Donato
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Paolo Zaffino
- Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Elisabetta Scali
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Krizia Caterina Pirrone
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Maria Francesca Spadea
- Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Natalia Malara
- Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Giuseppe Donato
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| |
Collapse
|
20
|
Montes Diaz G, Hupperts R, Fraussen J, Somers V. Dimethyl fumarate treatment in multiple sclerosis: Recent advances in clinical and immunological studies. Autoimmun Rev 2018; 17:1240-1250. [DOI: 10.1016/j.autrev.2018.07.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/11/2018] [Indexed: 12/30/2022]
|
21
|
Tahrali I, Kucuksezer UC, Altintas A, Uygunoglu U, Akdeniz N, Aktas-Cetin E, Deniz G. Dysfunction of CD3 -CD16 +CD56 dim and CD3 -CD16 -CD56 bright NK cell subsets in RR-MS patients. Clin Immunol 2018; 193:88-97. [PMID: 29448007 DOI: 10.1016/j.clim.2018.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 02/09/2018] [Accepted: 02/10/2018] [Indexed: 02/09/2023]
Affiliation(s)
- Ilhan Tahrali
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Umut Can Kucuksezer
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Ayse Altintas
- Istanbul University, Cerrahpasa Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| | - Ugur Uygunoglu
- Istanbul University, Cerrahpasa Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| | - Nilgun Akdeniz
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Esin Aktas-Cetin
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Gunnur Deniz
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey.
| |
Collapse
|
22
|
Dimethyl fumarate induces a persistent change in the composition of the innate and adaptive immune system in multiple sclerosis patients. Sci Rep 2018; 8:8194. [PMID: 29844361 PMCID: PMC5974280 DOI: 10.1038/s41598-018-26519-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/14/2018] [Indexed: 11/22/2022] Open
Abstract
The effects of dimethyl fumarate (DMF) on the immune system in multiple sclerosis (MS) are not completely elucidated. In this study, an extensive immunophenotypic analysis of innate and adaptive immune cells of DMF-treated MS patients was performed. Peripheral blood immune cell phenotypes were determined using flow cytometry in a follow-up study of 12 MS patients before, after 3 and 12 months of DMF treatment and a cross-sectional study of 25 untreated and 64 DMF-treated MS patients. Direct effects of DMF on B cells were analyzed in vitro. After 12 months of DMF treatment, percentages of monocytes, natural killer cells, naive T and B cells and transitional B cells increased. Percentages of (effector) memory T cells, (non) class-switched memory B cells and double negative B cells decreased together with CD4+ T cells expressing interferon-γ (IFN-γ), granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin-17 (IL-17). DMF treatment was fully effective as of 6 months and directly induced apoptosis and decreased expression of costimulatory CD40, antigen presentation molecule MHCII and B cell activating factor receptor (BAFFR) on B cells. DMF induced a persistent change of the immune system of MS patients, directly induced apoptosis and reduced expression of functional markers on B cells.
Collapse
|
23
|
Gianchecchi E, Delfino DV, Fierabracci A. NK cells in autoimmune diseases: Linking innate and adaptive immune responses. Autoimmun Rev 2018; 17:142-154. [PMID: 29180124 DOI: 10.1016/j.autrev.2017.11.018] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The pathogenesis of autoimmunity remains to be fully elucidated, although the contribution of genetic and environmental factors is generally recognized. Despite autoimmune conditions are principally due to T and B lymphocytes, NK cells also appear to play a role in the promotion and/or maintenance of altered adaptive immune responses or in peripheral tolerance mechanisms. Although NK cells are components of the innate immune system, they shows characteristics of the adaptive immune system, such as the expansion of pathogen-specific cells, the generation of long-lasting "memory" cells able to persist upon cognate antigen encounter, and the possibility to induce an increased secondary recall response to re-challenge. Human NK cells are generally identified as CD56+CD3-, conversely CD56+CD3+ cells represent a mixed population of NK-like T (NK T) cells and antigen-experienced T cells showing the up-regulation of several NK cell markers. CD56dim constitute about 90% of NK cells in the peripheral blood, they are mature and involved in cytotoxicity responses; CD56bright instead are more immature, mostly involved in cytokine production, having only a limited role in cytolytic responses, keen to leave the blood vessels as the principal population observed in lymph nodes. NK cells have been identified also in non-lymphoid tissues since, in pathologic conditions, they can quickly reach the target organs. A cross-talk between NK with dendritic cells and T cells is established throughout different receptor-ligand bindings. Several studies support the correlation between NK cell number and/or functional alterations, such as a defective cytotoxic activity and several autoimmune conditions. Among the different autoimmune pathologies and even within the same disease, NK cell function is significantly different either promoting or even protecting against the onset of the autoimmune condition. In this Review, we discuss recent literature supporting the role played by NK cells, as a bridge between innate and adaptive immunity, in the onset of autoimmune diseases.
Collapse
Affiliation(s)
- Elena Gianchecchi
- Type 1 Diabetes Centre, Infectivology and Clinical Trials Research Department, Children's Hospital Bambino Gesù, Rome, Italy
| | | | - Alessandra Fierabracci
- Type 1 Diabetes Centre, Infectivology and Clinical Trials Research Department, Children's Hospital Bambino Gesù, Rome, Italy.
| |
Collapse
|
24
|
Abstract
Multiple sclerosis (MS) is a chronic disease of the central nervous system (CNS) characterized by loss of motor and sensory function that results from immune-mediated inflammation, demyelination, and subsequent axonal damage. Clinically, most MS patients experience recurrent episodes (relapses) of neurological impairment, but in most cases (60–80%) the course of the disease eventually becomes chronic and progressive, leading to cumulative motor, sensory, and visual disability, and cognitive deficits. The course of the disease is largely unpredictable and its clinical presentation is variable, but its predilection for certain parts of the CNS, which includes the optic nerves, the brain stem, cerebellum, and cervical spinal cord, provides a characteristic constellation of signs and symptoms. Several variants of MS have been nowadays defined with variable immunopathogenesis, course and prognosis. Many new treatments targeting the immune system have shown efficacy in preventing the relapses of MS and have been introduced to its management during the last decade.
Collapse
|
25
|
Bernardini G, Antonangeli F, Bonanni V, Santoni A. Dysregulation of Chemokine/Chemokine Receptor Axes and NK Cell Tissue Localization during Diseases. Front Immunol 2016; 7:402. [PMID: 27766097 PMCID: PMC5052267 DOI: 10.3389/fimmu.2016.00402] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 09/21/2016] [Indexed: 01/06/2023] Open
Abstract
Chemokines are small chemotactic molecules that play key roles in physiological and pathological conditions. Upon signaling via their specific receptors, chemokines regulate tissue mobilization and trafficking of a wide array of immune cells, including natural killer (NK) cells. Current research is focused on analyzing changes in chemokine/chemokine receptor expression during various diseases to interfere with pathological trafficking of cells or to recruit selected cell types to specific tissues. NK cells are a heterogeneous lymphocyte population comprising several subsets endowed with distinct functional properties and mainly representing distinct stages of a linear development process. Because of their different functional potential, the type of subset that accumulates in a tissue drives the final outcome of NK cell-regulated immune response, leading to either protection or pathology. Correspondingly, chemokine receptors, including CXCR4, CXCR3, and CX3CR1, are differentially expressed by NK cell subsets, and their expression levels can be modulated during NK cell activation. At first, this review will summarize the current knowledge on the contribution of chemokines to the localization and generation of NK cell subsets in homeostasis. How an inappropriate chemotactic response can lead to pathology and how chemokine targeting can therapeutically affect tissue recruitment/localization of distinct NK cell subsets will also be discussed.
Collapse
Affiliation(s)
- Giovanni Bernardini
- Department of Molecular Medicine, Sapienza University, Rome, Italy; IRCCS NEUROMED - Mediterranean Neurological Institute, Isernia, Italy
| | | | - Valentina Bonanni
- Department of Molecular Medicine, Sapienza University , Rome , Italy
| | - Angela Santoni
- IRCCS NEUROMED - Mediterranean Neurological Institute, Isernia, Italy; Department of Molecular Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
26
|
Hertwig L, Hamann I, Romero-Suarez S, Millward JM, Pietrek R, Chanvillard C, Stuis H, Pollok K, Ransohoff RM, Cardona AE, Infante-Duarte C. CX3CR1-dependent recruitment of mature NK cells into the central nervous system contributes to control autoimmune neuroinflammation. Eur J Immunol 2016; 46:1984-96. [PMID: 27325505 DOI: 10.1002/eji.201546194] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/28/2016] [Accepted: 06/13/2016] [Indexed: 12/12/2022]
Abstract
Fractalkine receptor (CX3CR1)-deficient mice develop very severe experimental autoimmune encephalomyelitis (EAE), associated with impaired NK cell recruitment into the CNS. Yet, the precise implications of NK cells in autoimmune neuroinflammation remain elusive. Here, we investigated the pattern of NK cell mobilization and the contribution of CX3CR1 to NK cell dynamics in the EAE. We show that in both wild-type and CX3CR1-deficient EAE mice, NK cells are mobilized from the periphery and accumulate in the inflamed CNS. However, in CX3CR1-deficient mice, the infiltrated NK cells displayed an immature phenotype contrasting with the mature infiltrates in WT mice. This shift in the immature/mature CNS ratio contributes to EAE exacerbation in CX3CR1-deficient mice, since transfer of mature WT NK cells prior to immunization exerted a protective effect and normalized the CNS NK cell ratio. Moreover, mature CD11b(+) NK cells show higher degranulation in the presence of autoreactive 2D2 transgenic CD4(+) T cells and kill these autoreactive cells more efficiently than the immature CD11b(-) fraction. Together, these data suggest a protective role of mature NK cells in EAE, possibly through direct modulation of T cells inside the CNS, and demonstrate that mature and immature NK cells are recruited into the CNS by distinct chemotactic signals.
Collapse
Affiliation(s)
- Laura Hertwig
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, a joint cooperation between the Charité - Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Isabell Hamann
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, a joint cooperation between the Charité - Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Silvina Romero-Suarez
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, a joint cooperation between the Charité - Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Jason M Millward
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, a joint cooperation between the Charité - Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Rebekka Pietrek
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, a joint cooperation between the Charité - Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Coralie Chanvillard
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, a joint cooperation between the Charité - Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Hanna Stuis
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, a joint cooperation between the Charité - Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Karolin Pollok
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, a joint cooperation between the Charité - Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany.,German Rheumatism Research Center, Germany and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Astrid E Cardona
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Carmen Infante-Duarte
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, a joint cooperation between the Charité - Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
27
|
Huth TK, Brenu EW, Ramos S, Nguyen T, Broadley S, Staines D, Marshall-Gradisnik S. Pilot Study of Natural Killer Cells in Chronic Fatigue Syndrome/Myalgic Encephalomyelitis and Multiple Sclerosis. Scand J Immunol 2016; 83:44-51. [PMID: 26381393 DOI: 10.1111/sji.12388] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 09/05/2015] [Indexed: 12/27/2022]
Abstract
Patients with chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) and multiple sclerosis (MS) suffer from debilitating fatigue which is not alleviated by rest. In addition to the fatigue-related symptoms suffered by patients with CFS/ME and MS, dysfunction of the immune system and, in particular, reduced natural killer (NK) cell cytotoxic activity has also been reported in CFS/ME and MS. The purpose of this pilot study was to compare NK cellular mechanisms in patients with CFS/ME and MS to investigate potential dysfunctions in the NK cell activity pathway. Flow cytometry protocols assessed CD56(dim) CD16(+) and CD56(bright) CD16(+/-) NK cell expression of adhesion molecules, NK activating and inhibiting receptors, NK cell maturation and lytic proteins. All participants in this study were female and included 14 patients with CFS/ME, nine patients with MS and 19 non-fatigued controls. The patient groups and the non-fatigued controls were not taking any immunosuppressive or immune-enhancing medications. In the MS cohort, KIR2DL5 was significantly increased on CD56(bright) CD16(+/-) NK cells and expression of CD94 was significantly increased on CD56(dim) CD16(+) NK cells in comparison with the controls. Co-expression of CD57 and perforin was significantly increased on CD56(dim) CD16(+) NK cells from patients with CFS/ME compared to the MS and non-fatigued control participants. The results from this pilot study suggest that NK cells from patients with CFS/ME and MS may have undergone increased differentiation in response to external stimuli which may affect different mechanisms in the NK cell cytotoxic activity pathway.
Collapse
Affiliation(s)
- T K Huth
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Southport, Qld, Australia.,School of Medical Science, Griffith University, Southport, Qld, Australia
| | - E W Brenu
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Southport, Qld, Australia.,School of Medical Science, Griffith University, Southport, Qld, Australia
| | - S Ramos
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Southport, Qld, Australia.,School of Medical Science, Griffith University, Southport, Qld, Australia
| | - T Nguyen
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Southport, Qld, Australia.,School of Medical Science, Griffith University, Southport, Qld, Australia
| | - S Broadley
- School of Medicine, Griffith University, Southport, Qld, Australia.,Gold Coast University Hospital, Southport, Qld, Australia
| | - D Staines
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Southport, Qld, Australia.,School of Medical Science, Griffith University, Southport, Qld, Australia
| | - S Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Southport, Qld, Australia.,School of Medical Science, Griffith University, Southport, Qld, Australia
| |
Collapse
|
28
|
Gadani SP, Kipnis J. Natural killers in the brain's nursery. Nat Neurosci 2016; 19:176-7. [PMID: 26814583 DOI: 10.1038/nn.4227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sachin P Gadani
- Center for Brain Immunology and Glia, Department of Neuroscience, Graduate Program in Neuroscience and Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, Graduate Program in Neuroscience and Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
29
|
Edwards SC, McGinley AM, McGuinness NC, Mills KHG. γδ T Cells and NK Cells - Distinct Pathogenic Roles as Innate-Like Immune Cells in CNS Autoimmunity. Front Immunol 2015; 6:455. [PMID: 26441960 PMCID: PMC4561808 DOI: 10.3389/fimmu.2015.00455] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 08/24/2015] [Indexed: 12/31/2022] Open
Affiliation(s)
- Sarah C Edwards
- Immune Regulation Research Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin , Dublin , Ireland
| | - Aoife M McGinley
- Immune Regulation Research Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin , Dublin , Ireland
| | - Niamh C McGuinness
- Immune Regulation Research Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin , Dublin , Ireland ; Trinity College Institute of Neuroscience, Trinity College Dublin , Dublin , Ireland
| | - Kingston H G Mills
- Immune Regulation Research Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
30
|
Rodríguez-Martín E, Picón C, Costa-Frossard L, Alenda R, Sainz de la Maza S, Roldán E, Espiño M, Villar LM, Álvarez-Cermeño JC. Natural killer cell subsets in cerebrospinal fluid of patients with multiple sclerosis. Clin Exp Immunol 2015; 180:243-9. [PMID: 25565222 DOI: 10.1111/cei.12580] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/25/2014] [Indexed: 01/12/2023] Open
Abstract
Changes in blood natural killer (NK) cells, important players of the immune innate system, have been described in multiple sclerosis (MS). We studied percentages and total cell counts of different effector and regulatory NK cells in cerebrospinal fluid (CSF) of MS patients and other neurological diseases to gain clearer knowledge of the role of these cells in neuroinflammation. NK cell subsets were assessed by flow cytometry in CSF of 85 consecutive MS patients (33 with active disease and 52 with stable MS), 16 with other inflammatory diseases of the central nervous system (IND) and 17 with non-inflammatory neurological diseases (NIND). MS patients showed a decrease in percentages of different CSF NK subpopulations compared to the NIND group. However, absolute cell counts showed a significant increase of all NK subsets in MS and IND patients, revealing that the decrease in percentages does not reflect a real reduction of these immune cells. Remarkably, MS patients showed a significant increase of regulatory/effector (CD56(bright) /CD56(dim) ) NK ratio compared to IND and NIND groups. In addition, MS activity associated with an expansion of NK T cells. These data show that NK cell subsets do not increase uniformly in all inflammatory neurological disease and suggest strongly that regulatory CD56(bright) and NK T cells may arise in CSF of MS patients as an attempt to counteract the CNS immune activation characteristic of the disease.
Collapse
Affiliation(s)
- E Rodríguez-Martín
- Department of Immunology, Multiple Sclerosis Unit, Hospital Ramón y Cajal, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria IRYCIS, Madrid, Spain; Red Española de Esclerosis Múltiple, REEM, Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Serrano-Pertierra E, Blanco-Gelaz MA, Oliva-Nacarino P, Martínez-Camblor P, Villafani J, López-Larrea C, Cernuda-Morollón E. Increased natural killer cell chemotaxis to CXCL12 in patients with multiple sclerosis. J Neuroimmunol 2015; 282:39-44. [PMID: 25903727 DOI: 10.1016/j.jneuroim.2015.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/04/2015] [Accepted: 03/06/2015] [Indexed: 12/17/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory and neurodegenerative disease characterized by leukocyte infiltration into the central nervous system (CNS). Migration of lymphocyte subpopulations towards CXCL12 was analyzed coupled to six-color flow cytometry in untreated patients in the remitting phase, during relapse, in patients with clinically isolated syndrome (CIS), and in healthy volunteers. Significantly higher migration rates of natural killer cells (CD45+CD3-CD16/56+) were observed in patients in remission and CIS patients than in patients during relapse and in controls. Moreover, the frequency of CD3-CD16/56+CXCR4+ cells is higher in patients in remission and in CIS patients, but not during relapse.
Collapse
Affiliation(s)
- Esther Serrano-Pertierra
- Immunology Department, Hospital Universitario Central de Asturias, Oviedo 33011, Spain; Neurology Department, Hospital Universitario Central de Asturias, Oviedo 33011, Spain.
| | | | - Pedro Oliva-Nacarino
- Neurology Department, Hospital Universitario Central de Asturias, Oviedo 33011, Spain.
| | - Pablo Martínez-Camblor
- Oficina de Investigación Sanitaria, Asturias, Spain; Universidad Autónoma de Chile, Chile.
| | - Javier Villafani
- Neurology Department, Hospital Universitario Central de Asturias, Oviedo 33011, Spain.
| | - Carlos López-Larrea
- Immunology Department, Hospital Universitario Central de Asturias, Oviedo 33011, Spain; Fundación Renal Íñigo Álvarez de Toledo, Madrid 28003, Spain.
| | - Eva Cernuda-Morollón
- Neurology Department, Hospital Universitario Central de Asturias, Oviedo 33011, Spain.
| |
Collapse
|
32
|
Ellwardt E, Zipp F. Molecular mechanisms linking neuroinflammation and neurodegeneration in MS. Exp Neurol 2014; 262 Pt A:8-17. [DOI: 10.1016/j.expneurol.2014.02.006] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/31/2014] [Accepted: 02/07/2014] [Indexed: 12/21/2022]
|
33
|
Abstract
BACKGROUND Many aspects of autoimmune disease are not well understood, including the specificities of autoimmune targets, and patterns of co-morbidity and cross-heritability across diseases. Prior work has provided evidence that somatic mutation caused by gene conversion and deletion at segmentally duplicated loci is relevant to several diseases. Simple tandem repeat (STR) sequence is highly mutable, both somatically and in the germ-line, and somatic STR mutations are observed under inflammation. RESULTS Protein-coding genes spanning STRs having markers of mutability, including germ-line variability, high total length, repeat count and/or repeat similarity, are evaluated in the context of autoimmunity. For the initiation of autoimmune disease, antigens whose autoantibodies are the first observed in a disease, termed primary autoantigens, are informative. Three primary autoantigens, thyroid peroxidase (TPO), phogrin (PTPRN2) and filaggrin (FLG), include STRs that are among the eleven longest STRs spanned by protein-coding genes. This association of primary autoantigens with long STR sequence is highly significant (p<3.0x10(-7)). Long STRs occur within twenty genes that are associated with sixteen common autoimmune diseases and atherosclerosis. The repeat within the TTC34 gene is an outlier in terms of length and a link with systemic lupus erythematosus is proposed. CONCLUSIONS The results support the hypothesis that many autoimmune diseases are triggered by immune responses to proteins whose DNA sequence mutates somatically in a coherent, consistent fashion. Other autoimmune diseases may be caused by coherent somatic mutations in immune cells. The coherent somatic mutation hypothesis has the potential to be a comprehensive explanation for the initiation of many autoimmune diseases.
Collapse
Affiliation(s)
- Kenneth Andrew Ross
- Department of Computer Science, Columbia University, New York, New York, United States of America
| |
Collapse
|
34
|
Sheng W, Liu C, Fu R, Wang H, Qu W, Ruan E, Wang G, Liu H, Wu Y, Song J, Xing L, Guan J, Li L, Liu H, Shao Z. Abnormalities of quantities and functions of linker for activations of T cells in severe aplastic anemia. Eur J Haematol 2014; 93:214-23. [PMID: 24673455 DOI: 10.1111/ejh.12327] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2014] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Severe aplastic anemia (SAA) is a rare immune-regulated disease characterized by severe pancytopenia and bone marrow failure, caused by destruction of hematopoietic cells by the activated T lymphocytes. Linker for activation of T cells (LAT), a transmembrane adaptor protein, plays a key role in T-cell and mast cell functions. However, it remains unclear how LAT may change in patients with SAA. This study aims at understanding the role of lymphocyte LAT in SAA. METHODS The expression of LAT, related signaling molecules, and T-cell effector molecules was determined by flow cytometry. LAT mRNA was evaluated by quantitative real-time PCR. Cytokine production by cultured T cells was determined by ELISA. RESULTS Patients with SAA had an increased levels of LAT and both total phosphorylated LAT and of the related molecule (ZAP-70) in circulating T cells compared with normal controls. In patients with SAA, the expression of LAT was positively associated with the expression of perforin and granzyme B in CD8(+) T cells. Inhibition of LAT expression in T cells from patients with SAA decreased the activation of the CD4(+) and CD8(+) T-cell subsets. Overexpression of LAT in T cells from normal controls increased the activation of CD4(+) and CD8(+) T-cell subsets with increased apoptosis of K562 cells in coculture. CONCLUSIONS Our findings demonstrate that dysregulation of LAT expression and activation may contribute to over-function of T cells, imbalance of Th1/Th2 subsets and thus lead to hematopoiesis failure in SAA. Immunosuppressive therapy dramatically reduced the expression of LAT making it an attractive therapeutic target in SAA.
Collapse
Affiliation(s)
- Weiwei Sheng
- The Department of Hematology, The General Hospital of Tianjin Medical University, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Liu C, Li Z, Sheng W, Fu R, Li L, Zhang T, Wu Y, Xing L, Song J, Wang H, Shao Z. Abnormalities of quantities and functions of natural killer cells in severe aplastic anemia. Immunol Invest 2014; 43:491-503. [PMID: 24661133 DOI: 10.3109/08820139.2014.888448] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Severe aplastic anemia (SAA) is a rare disease characterized by severe pancytopenia and bone marrow failure. Natural killer (NK) cells are large granular lymphocytes derived from hematopoietic stem cells (HSCs) or common lymphoid progenitors (CLP). They play a key role in n the innate immunity and adaptive immune. In this study, the quantitative and functional changes of natural killer (NK) cell subsets in peripheral blood of severe aplastic anemia (SAA) patients before and after immunosuppressive therapy (IST) were investigated. Results showed that the percentage of NK cells and its subsets in peripheral blood lymphocytes was decreased in SAA patients. After IST, the percentage of NK cells and their subsets increased dramatically. The median expressions of CD158a, NKG2D and NKp46 on NK cells were higher in SAA patients compared to that in normal controls, and the expressions of perforin in newly diagnosed and recovery SAA patients were higher than that in controls. Therefore, we concluded that the decrease of total NK cells, and CD56(bright), CD56(dim) NK cell subsets and the higher expressions of NKp46 and perforin on NK cells may cause the over-function of T lymphocytes and thus lead to hematopoiesis failure in SAA.
Collapse
Affiliation(s)
- Chunyan Liu
- Department of Hematology, General Hospital of Tianjin Medical University , Tianjin , P.R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Poggi A, Zocchi MR. NK cell autoreactivity and autoimmune diseases. Front Immunol 2014; 5:27. [PMID: 24550913 PMCID: PMC3912987 DOI: 10.3389/fimmu.2014.00027] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 01/17/2014] [Indexed: 01/14/2023] Open
Abstract
Increasing evidences have pointed out the relevance of natural killer (NK) cells in organ-specific and systemic autoimmune diseases. NK cells bear a plethora of activating and inhibiting receptors that can play a role in regulating reactivity with autologous cells. The activating receptors recognize natural ligands up-regulated on virus-infected or stressed or neoplastic cells. Of note, several autoimmune diseases are thought to be linked to viral infections as one of the first event in inducing autoimmunity. Also, it is conceivable that autoimmunity can be triggered when a dysregulation of innate immunity occurs, activating T and B lymphocytes to react with self-components. This would imply that NK cells can play a regulatory role during adaptive immunity; indeed, innate lymphoid cells (ILCs), comprising the classical CD56(+) NK cells, have a role in maintaining or alternating tissue homeostasis secreting protective and/or pro-inflammatory cytokines. In addition, NK cells display activating receptors involved in natural cytotoxicity and the activating isoforms of receptors for HLA class I that can interact with healthy host cells and induce damage without any evidence of viral infection or neoplastic-induced alteration. In this context, the interrelationship among ILC, extracellular-matrix components, and mesenchymal stromal cells can be considered a key point for the control of homeostasis. Herein, we summarize evidences for a role of NK cells in autoimmune diseases and will give a point of view of the interplay between NK cells and self-cells in triggering autoimmunity.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, Scientific Institute San Raffaele, Milan, Italy
| |
Collapse
|
37
|
Morris G, Maes M. Myalgic encephalomyelitis/chronic fatigue syndrome and encephalomyelitis disseminata/multiple sclerosis show remarkable levels of similarity in phenomenology and neuroimmune characteristics. BMC Med 2013; 11:205. [PMID: 24229326 PMCID: PMC3847236 DOI: 10.1186/1741-7015-11-205] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 08/15/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND 'Encephalomyelitis disseminata' (multiple sclerosis) and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) are both classified as diseases of the central nervous system by the World Health Organization. This review aims to compare the phenomenological and neuroimmune characteristics of MS with those of ME/CFS. DISCUSSION There are remarkable phenomenological and neuroimmune overlaps between both disorders. Patients with ME/CFS and MS both experience severe levels of disabling fatigue and a worsening of symptoms following exercise and resort to energy conservation strategies in an attempt to meet the energy demands of day-to-day living. Debilitating autonomic symptoms, diminished cardiac responses to exercise, orthostatic intolerance and postural hypotension are experienced by patients with both illnesses. Both disorders show a relapsing-remitting or progressive course, while infections and psychosocial stress play a large part in worsening of fatigue symptoms. Activated immunoinflammatory, oxidative and nitrosative (O+NS) pathways and autoimmunity occur in both illnesses. The consequences of O+NS damage to self-epitopes is evidenced by the almost bewildering and almost identical array of autoantibodies formed against damaged epitopes seen in both illnesses. Mitochondrial dysfunctions, including lowered levels of ATP, decreased phosphocreatine synthesis and impaired oxidative phosphorylation, are heavily involved in the pathophysiology of both MS and ME/CFS. The findings produced by neuroimaging techniques are quite similar in both illnesses and show decreased cerebral blood flow, atrophy, gray matter reduction, white matter hyperintensities, increased cerebral lactate and choline signaling and lowered acetyl-aspartate levels. SUMMARY This review shows that there are neuroimmune similarities between MS and ME/CFS. This further substantiates the view that ME/CFS is a neuroimmune illness and that patients with MS are immunologically primed to develop symptoms of ME/CFS.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Pembrey, Llanelli, UK
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - Michael Maes
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
- Department of Psychiatry, Deakin University, Geelong, Australia
| |
Collapse
|
38
|
Chanvillard C, Jacolik RF, Infante-Duarte C, Nayak RC. The role of natural killer cells in multiple sclerosis and their therapeutic implications. Front Immunol 2013; 4:63. [PMID: 23493880 PMCID: PMC3595639 DOI: 10.3389/fimmu.2013.00063] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 02/27/2013] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is assumed to be an autoimmune disease initiated by autoreactive T cells that recognize central nervous system antigens. Although adaptive immunity is clearly involved in MS pathogenesis, innate immunity increasingly appears to be implicated in the disease. We and others have presented evidence that natural killer (NK) cells may be involved in immunoregulation in MS, leading to the question of whether a particular NK cell subtype will account for this effect. Changes of NK cell functionality in MS were associated with MS activity, and depletion of NK cells exacerbated the course of disease in a murine model of MS, experimental autoimmune encephalomyelitis. Several studies described a deficiency and transient "valleys" in NK cell killing activity in human MS, which may coincide with symptomatic relapse. However, the molecular basis of the defect in killing activity has not been determined. We discuss results on the expression of perforin in CD16(+) NK cells and the existence of an inverse relationship between myelin loaded phagocytes and the proportion of CD16(+) NK cells expressing perforin in the circulation. This inverse relationship is consistent with a role for NK cell killing activity in dampening autoimmunity. On the other hand, it has been broadly reported that first line MS therapies, such as interferon-beta, glatiramer acetate as well as escalation therapies such as fingolimod, daclizumab, or mitoxantrone seem to affect NK cell functionality and phenotype in vivo. Therefore, in this review we consider evidence for the immunoregulatory role of NK cells in MS and its animal models. Furthermore, we discuss the effect of MS treatments on NK cell activity.
Collapse
Affiliation(s)
- Coralie Chanvillard
- Institute of Medical Immunology, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, A Joint Cooperation Between the Charité, Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine Berlin, Germany
| | | | | | | |
Collapse
|
39
|
Durrenberger PF, Webb LV, Sim MJW, Nicholas RS, Altmann DM, Boyton RJ. Increased HLA-E expression in white matter lesions in multiple sclerosis. Immunology 2012; 137:317-25. [PMID: 23039207 PMCID: PMC3530087 DOI: 10.1111/imm.12012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 07/22/2012] [Accepted: 09/11/2012] [Indexed: 12/24/2022] Open
Abstract
The molecular mechanisms underpinning central nervous system damage in multiple sclerosis (MS) are complex and it is widely accepted that there is an autoimmune component. Both adaptive and innate immune effector mechanisms are believed to contribute to tissue disease aetiology. HLA-E is a non-classical MHC class Ib molecule that acts as the ligand for the NKG2A inhibitory receptor present on natural killer (NK) and CD8+ cells. Peptide binding and stabilization of HLA-E is often considered to signal infection or cell stress. Here we examine the up-regulation of HLA-E in MS brain tissue. Expression is significantly increased in white matter lesions in the brain of MS patients compared with white matter of neurologically healthy controls. Furthermore, using quantitative immunohistochemistry and confocal microscopy, we show increased HLA-E protein expression in endothelial cells of active MS lesions. Non-inflammatory chronic lesions express significantly less HLA-E protein, comparable to levels found in white matter from controls. Increased HLA-E protein levels were associated with higher scores of inflammation. These results suggest the potential for an effect in central nervous system pathogenesis from HLA-E modulation in stressed tissue. Co-localization with infiltrating CD8+ cells implicates a possible role for HLA-E-restricted regulatory CD8+ cells, as has been proposed in other autoimmune diseases.
Collapse
Affiliation(s)
- Pascal F Durrenberger
- Department of Medicine, Section of Infectious Diseases and Immunity, Hammersmith Hospital, Imperial College, London, UK
| | | | | | | | | | | |
Collapse
|
40
|
Kaur G, Trowsdale J, Fugger L. Natural killer cells and their receptors in multiple sclerosis. ACTA ACUST UNITED AC 2012; 136:2657-76. [PMID: 22734127 DOI: 10.1093/brain/aws159] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system has crucial roles in the pathogenesis of multiple sclerosis. While the adaptive immune cell subsets, T and B cells, have been the main focus of immunological research in multiple sclerosis, it is now important to realize that the innate immune system also has a key involvement in regulating autoimmune responses in the central nervous system. Natural killer cells are innate lymphocytes that play vital roles in a diverse range of infections. There is evidence that they influence a number of autoimmune conditions. Recent studies in multiple sclerosis and its murine model, experimental autoimmune encephalomyelitis, are starting to provide some understanding of the role of natural killer cells in regulating inflammation in the central nervous system. Natural killer cells express a diverse range of polymorphic cell surface receptors, which interact with polymorphic ligands; this interaction controls the function and the activation status of the natural killer cell. In this review, we discuss evidence for the role of natural killer cells in multiple sclerosis and experimental autoimmune encephalomyelitis. We consider how a change in the balance of signals received by the natural killer cell influences its involvement in the ensuing immune response, in relation to multiple sclerosis.
Collapse
Affiliation(s)
- Gurman Kaur
- MRC Human Immunology Unit, Nuffield Department of Medicine, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | | | | |
Collapse
|
41
|
Cytotoxicity of CD56(bright) NK cells towards autologous activated CD4+ T cells is mediated through NKG2D, LFA-1 and TRAIL and dampened via CD94/NKG2A. PLoS One 2012; 7:e31959. [PMID: 22384114 PMCID: PMC3284517 DOI: 10.1371/journal.pone.0031959] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 01/16/2012] [Indexed: 12/16/2022] Open
Abstract
In mouse models of chronic inflammatory diseases, Natural Killer (NK) cells can play an immunoregulatory role by eliminating chronically activated leukocytes. Indirect evidence suggests that NK cells may also be immunoregulatory in humans. Two subsets of human NK cells can be phenotypically distinguished as CD16+CD56dim and CD16dim/−CD56bright. An expansion in the CD56bright NK cell subset has been associated with clinical responses to therapy in various autoimmune diseases, suggesting an immunoregulatory role for this subset in vivo. Here we compared the regulation of activated human CD4+ T cells by CD56dim and CD56bright autologous NK cells in vitro. Both subsets efficiently killed activated, but not resting, CD4+ T cells. The activating receptor NKG2D, as well as the integrin LFA-1 and the TRAIL pathway, played important roles in this process. Degranulation by NK cells towards activated CD4+ T cells was enhanced by IL-2, IL-15, IL-12+IL-18 and IFN-α. Interestingly, IL-7 and IL-21 stimulated degranulation by CD56bright NK cells but not by CD56dim NK cells. NK cell killing of activated CD4+ T cells was suppressed by HLA-E on CD4+ T cells, as blocking the interaction between HLA-E and the inhibitory CD94/NKG2A NK cell receptor enhanced NK cell degranulation. This study provides new insight into CD56dim and CD56bright NK cell-mediated elimination of activated autologous CD4+ T cells, which potentially may provide an opportunity for therapeutic treatment of chronic inflammation.
Collapse
|
42
|
Harrer A, Pilz G, Einhaeupl M, Oppermann K, Hitzl W, Wipfler P, Sellner J, Golaszewski S, Afazel S, Haschke-Becher E, Trinka E, Kraus J. Lymphocyte subsets show different response patterns to in vivo bound natalizumab--a flow cytometric study on patients with multiple sclerosis. PLoS One 2012; 7:e31784. [PMID: 22363732 PMCID: PMC3282779 DOI: 10.1371/journal.pone.0031784] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 01/17/2012] [Indexed: 11/18/2022] Open
Abstract
Natalizumab is an effective monoclonal antibody therapy for the treatment of relapsing-remitting multiple sclerosis (RRMS) and interferes with immune cell migration into the central nervous system by blocking the α(4) subunit of very-late activation antigen-4 (VLA-4). Although well tolerated and very effective, some patients still suffer from relapses in spite of natalizumab therapy or from unwanted side effects like progressive multifocal leukoencephalopathy (PML). In search of a routine-qualified biomarker on the effectiveness of natalizumab therapy we applied flow cytometry and analyzed natalizumab binding to α(4) and α(4) integrin surface levels on T-cells, B-cells, natural killer (NK) cells, and NKT cells from 26 RRMS patients under up to 72 weeks of therapy. Four-weekly infusions of natalizumab resulted in a significant and sustained increase of lymphocyte-bound natalizumab (p<0.001) which was paralleled by a significant decrease in detectability of the α(4) integrin subunit on all lymphocyte subsets (p<0.001). We observed pronounced natalizumab accumulations on T and B cells at single measurements in all patients who reported clinical disease activity (n = 4). The natalizumab binding capacity of in vitro saturated lymphocytes collected during therapy was strongly diminished compared to treatment-naive cells indicating a therapy-induced reduction of α(4). Summing up, this pilot study shows that flow cytometry is a useful method to monitor natalizumab binding to lymphocytes from RRMS patients under therapy. Investigating natalizumab binding provides an opportunity to evaluate the molecular level of effectiveness of natalizumab therapy in individual patients. In combination with natalizumab saturation experiments, it possibly even provides a means of studying the feasability of patient-tailored infusion intervals. A routine-qualified biomarker on the basis of individual natalizumab saturation on lymphocyte subsets might be an effective tool to improve treatment safety.
Collapse
Affiliation(s)
- Andrea Harrer
- Department of Neurology, Christian-Doppler-Klinik, Paracelsus Medical University, Salzburg, Austria.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Huarte E, Rynda-Apple A, Riccardi C, Skyberg JA, Golden S, Rollins MF, Ramstead AG, Jackiw LO, Maddaloni M, Pascual DW. Tolerogen-induced interferon-producing killer dendritic cells (IKDCs) protect against EAE. J Autoimmun 2011; 37:328-41. [PMID: 22018711 DOI: 10.1016/j.jaut.2011.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 09/13/2011] [Accepted: 09/17/2011] [Indexed: 11/15/2022]
Abstract
Natural killer (NK) cells and dendritic cells (DCs) have been shown to link the innate and adaptive immune systems. Likewise, a new innate cell subset, interferon-producing killer DCs (IKDCs), shares phenotypic and functional characteristics with both DCs and NK cells. Here, we show IKDCs play an essential role in the resolution of experimental autoimmune encephalomyelitis (EAE) upon treatment with the tolerizing agent, myelin oligodendrocyte glycoprotein (MOG), genetically fused to reovirus protein σ1 (termed MOG-pσ1). Activated IKDCs were recruited subsequent MOG-pσ1 treatment of EAE, and disease resolution was abated upon NK1.1 cell depletion. These IKDCs were able to kill activated CD4(+) T cells and mature dendritic DCs, thus, contributing to EAE remission. In addition, IKDCs were responsible for MOG-pσ1-mediated MOG-specific regulatory T cell recruitment to the CNS. The IKDCs induced by MOG-pσ1 expressed elevated levels of HVEM for interactions with cognate ligand-positive cells: LIGHT(+) NK and T(eff) cells and BTLA(+) B cells. Further characterization revealed these activated IKDCs being MHC class II(high), and upon their adoptive transfer (CD11c(+)NK1.1(+)MHC class II(high)), IKDCs, but not CD11c(+)NK1.1(+)MHC class II(intermediate/low) (unactivated) cells, conferred protection against EAE. These activated IKDCs showed enhanced CD107a, PD-L1, and granzyme B expression and could present OVA, unlike unactivated IKDCs. Thus, these results demonstrate the interventional potency induced HVEM(+) IKDCs to resolve autoimmune disease.
Collapse
Affiliation(s)
- Eduardo Huarte
- Department of Immunology and Infectious Diseases, Montana State University, 960 Technology Blvd., Bozeman, MT 59718, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Jiang W, Chai NR, Maric D, Bielekova B. Unexpected role for granzyme K in CD56bright NK cell-mediated immunoregulation of multiple sclerosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:781-90. [PMID: 21666061 PMCID: PMC3131478 DOI: 10.4049/jimmunol.1100789] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Functional NK cell deficiencies are associated with autoimmune diseases, including multiple sclerosis. NK cells can promote or inhibit adaptive immunity via either cytokine production or cytotoxicity toward immature dendritic cells and activated T cells. In humans, this immunoregulatory role resides in the CD56(bright) NK cell subset, which is selectively expanded by daclizumab, a CD25-blocking Ab that suppresses multiple sclerosis-associated inflammation. The objective of this study was to investigate the molecular mechanisms underlying the cytotoxicity of NK cells toward activated T cells. We demonstrated that NK cells induce caspase-independent apoptosis that requires NK cell degranulation and causes mitochondrial dysfunction in activated T cells. Although both granzyme A and granzyme K (GrK) can mediate this form of apoptosis, quantitatively we observed preferential transfer of GrK to target cells. Consequently, gene silencing of GrK in the NK-92 cell line, which retains functional characteristics of CD56(bright) NK cells, profoundly inhibited the ability of NK-92 cells to kill activated syngeneic T cells. Finally, we demonstrated that daclizumab treatment significantly enhanced this newly defined mechanism of cytotoxicity by CD56(bright) NK cells. Our study describes the important physiological role that GrK plays in immunoregulation of adaptive immunity in humans and indicates that therapeutic exploitation of this pathway is beneficial in controlling autoimmunity.
Collapse
Affiliation(s)
- Wenzheng Jiang
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- School of Life Science, East China Normal University, Shanghai 200062, China
| | - Noo Ri Chai
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dragan Maric
- Flow Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bibiana Bielekova
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
45
|
Sanvito L, Tomita A, Chihara N, Okamoto T, Lin Y, Ogawa M, Gran B, Aranami T, Yamamura T. Increase of Ki-67+ natural killer cells in multiple sclerosis patients treated with interferon-β and interferon-β combined with low-dose oral steroids. J Neuroimmunol 2011; 236:111-7. [PMID: 21652010 DOI: 10.1016/j.jneuroim.2011.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Revised: 04/26/2011] [Accepted: 05/11/2011] [Indexed: 01/21/2023]
Abstract
Interferon-β (IFN-β) is known to expand regulatory CD56(bright) natural killer (NK) cells in multiple sclerosis (MS). In this cross-sectional study we show that MS patients treated with IFN-β alone or in combination with low-dose prednisolone displayed increased proportion of all NK cell subsets in the active phase of the cell cycle (Ki-67+). There was no difference in NK cell apoptosis markers. In vitro experiments showed that both IFN-β and IFN-β in combination with corticosteroids increased the proportion of Ki-67(+) NK cells. This study, although limited, shows that treatment with IFN-β affects NK cell cycle without altering NK cell apoptosis in MS patients.
Collapse
Affiliation(s)
- Lara Sanvito
- Division of Clinical Neurology, University of Nottingham, Nottingham, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Graber JJ, Dhib-Jalbut S. Biomarkers of disease activity in multiple sclerosis. J Neurol Sci 2011; 305:1-10. [DOI: 10.1016/j.jns.2011.03.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 02/25/2011] [Accepted: 03/01/2011] [Indexed: 12/15/2022]
|
47
|
Lünemann A, Tackenberg B, DeAngelis T, da Silva RB, Messmer B, Vanoaica LD, Miller A, Apatoff B, Lublin FD, Lünemann JD, Münz C. Impaired IFN-γ production and proliferation of NK cells in multiple sclerosis. Int Immunol 2011; 23:139-48. [PMID: 21212154 PMCID: PMC3030728 DOI: 10.1093/intimm/dxq463] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 11/29/2010] [Indexed: 12/29/2022] Open
Abstract
NK cells are multicompetent lymphocytes of the innate immune system with a central role in host defense and immune regulation. Studies in experimental animal models of multiple sclerosis (MS) provided evidence for both pathologic and protective effects of NK cells. Humans harbor two functionally distinct NK-cell subsets exerting either predominantly cytotoxic (CD56(dim)CD16(+)) or immunoregulatory (CD56(bright)CD16(-)) functions. We analyzed these two subsets and their functions in the peripheral blood of untreated patients with relapsing-remitting MS compared with healthy blood donors. While ex vivo frequencies of CD56(bright)CD16(-) and CD56(dim)CD16(+) NK cells were similar in patients and controls, we found that cytokine-driven in vitro accumulation and IFN-γ production of CD56(bright)CD16(-) NK cells but not of their CD56(dim)CD16(+) counterparts were substantially diminished in MS. Impaired expansion of CD56(bright)CD16(-) NK cells was cell intrinsic because the observed effects could be reproduced with purified NK cells in an independent cohort of patients and controls. In contrast, cytolytic NK-cell activity toward the human erythromyeloblastoid leukemia cell line K562, the allogeneic CD4(+) T cell line CEM and allogeneic primary CD4(+) T-cell blasts was unchanged. Thus, characteristic functions of CD56(bright)CD16(-) NK cells, namely cytokine-induced NK cell expansion and IFN-γ production, are compromised in the NK cell compartment of MS patients.
Collapse
Affiliation(s)
- Anna Lünemann
- Christopher H. Browne Center for Immunology and Immune Diseases, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Martin JF, Perry JSA, Jakhete NR, Wang X, Bielekova B. An IL-2 paradox: blocking CD25 on T cells induces IL-2-driven activation of CD56(bright) NK cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:1311-20. [PMID: 20543101 PMCID: PMC3085179 DOI: 10.4049/jimmunol.0902238] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Daclizumab (Dac), an Ab against the IL-2R alpha-chain, inhibits brain inflammation in patients with multiple sclerosis, while expanding CD56(bright) immunoregulatory NK cells in vivo. We hypothesized that this unexpected expansion is paradoxically IL-2 driven; caused by the increased availability of T cell-derived IL-2 for NK cell signaling. To this end, we performed ex vivo functional analyses of CD56(bright) NK cells and T cells from patients in clinical trials with Dac. We developed in vitro models to investigate mechanisms for ex vivo observations. We observed that Dac treatment caused decreased numbers and proliferation of FoxP3(+) T regulatory cells (Tregs), a model T cell population known to be dependent on IL-2 for proliferation and survival. As anticipated, Dac therapy inhibited IL-2 signaling in all T cells; however, we also observed functional adaptation of T cells to low IL-2 signal in vivo, characterized by the concomitant enhancement of IL-7 signaling on all T cells and parallel increase of CD127 expression by Tregs. In contrast, IL-2 signaling on CD56(bright) NK cells was not inhibited by Dac and their in vivo proliferation and cytotoxicity actually increased. Mechanistic studies indicated that the activation of CD56(bright) NK cells was likely IL-2 driven, as low doses of IL-2, but not IL-15, mimicked this activation in vitro. Our study provides insight into the role that IL-2 and CD25 play in functional regulation of two important immunoregulatory cell populations in humans: FoxP3(+) Tregs and CD56(bright) NK cells.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- CD56 Antigen/immunology
- CD56 Antigen/metabolism
- Cell Line
- Cell Proliferation/drug effects
- Cells, Cultured
- Cytotoxicity, Immunologic/drug effects
- Cytotoxicity, Immunologic/immunology
- Daclizumab
- Flow Cytometry
- Forkhead Transcription Factors/immunology
- Forkhead Transcription Factors/metabolism
- Humans
- Immunoglobulin G/pharmacology
- Immunosuppressive Agents/pharmacology
- Interleukin-2/immunology
- Interleukin-2/metabolism
- Interleukin-2/pharmacology
- Interleukin-2 Receptor alpha Subunit/immunology
- Interleukin-2 Receptor alpha Subunit/metabolism
- Interleukin-7/immunology
- Interleukin-7/metabolism
- Interleukin-7 Receptor alpha Subunit/immunology
- Interleukin-7 Receptor alpha Subunit/metabolism
- K562 Cells
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Lewis X Antigen/immunology
- Lewis X Antigen/metabolism
- Lewis X Antigen/pharmacology
- Signal Transduction/drug effects
- Signal Transduction/immunology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Jayne F Martin
- Neuroimmunology Branch, National Institute for Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
49
|
Ramagopalan SV, Dobson R, Meier UC, Giovannoni G. Multiple sclerosis: risk factors, prodromes, and potential causal pathways. Lancet Neurol 2010; 9:727-39. [PMID: 20610348 DOI: 10.1016/s1474-4422(10)70094-6] [Citation(s) in RCA: 347] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a common, complex neurological disease. The precise aetiology of MS is not yet known, although epidemiological data indicate that both genetic and environmental factors are important. The evidence that the environment acts long before MS becomes clinically evident is well established and suggests the existence of a prodromal phase for the disease. The increasing incidence of MS emphasises the need for strategies to prevent this chronic disorder, and the possibility of a prodrome indicates a window of opportunity to potentially reverse early disease processes before clinical disease becomes evident. Studying a prodrome requires techniques other than clinical observation such as monitoring endophenotypes that result from associated risk factors. However, our current knowledge of causal pathways and endophenotypes in MS is limited. Identifying and studying individuals with a high risk of developing the disease provides a powerful opportunity to understand the MS causal cascade and is highly relevant to strategies that are aimed at preventing this debilitating disease.
Collapse
Affiliation(s)
- Sreeram V Ramagopalan
- Blizard Institute of Cell and Molecular Science, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
| | | | | | | |
Collapse
|
50
|
Role of the innate immune system in the pathogenesis of multiple sclerosis. J Neuroimmunol 2010; 221:7-14. [PMID: 19931190 DOI: 10.1016/j.jneuroim.2009.10.015] [Citation(s) in RCA: 226] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Revised: 10/05/2009] [Accepted: 10/06/2009] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease with heterogeneous clinical presentations and course. MS is considered to be a T cell mediated disease but in recent years contribution of innate immune cells in mediating MS pathogenesis is being appreciated. In this review, we have discussed the role of various innate immune cells in mediating MS. In particular, we have provided an overview of potential anti-inflammatory or pro-inflammatory function of DCs, microglial Cells, NK cells, NK-T cells and gamma delta T cells along with their interaction among themselves and with myelin. Given the understanding of the role of the innate immune cells in MS, it is possible that immunotherapeutic intervention targeting these cells may provide a better and effective treatment.
Collapse
|