1
|
Holicek P, Guilbaud E, Klapp V, Truxova I, Spisek R, Galluzzi L, Fucikova J. Type I interferon and cancer. Immunol Rev 2024; 321:115-127. [PMID: 37667466 DOI: 10.1111/imr.13272] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Type I interferon (IFN) is a class of proinflammatory cytokines with a dual role on malignant transformation, tumor progression, and response to therapy. On the one hand, robust, acute, and resolving type I IFN responses have been shown to mediate prominent anticancer effects, reflecting not only their direct cytostatic/cytotoxic activity on (at least some) malignant cells, but also their pronounced immunostimulatory functions. In line with this notion, type I IFN signaling has been implicated in the antineoplastic effects of various immunogenic therapeutics, including (but not limited to) immunogenic cell death (ICD)-inducing agents and immune checkpoint inhibitors (ICIs). On the other hand, weak, indolent, and non-resolving type I IFN responses have been demonstrated to support tumor progression and resistance to therapy, reflecting the ability of suboptimal type I IFN signaling to mediate cytoprotective activity, promote stemness, favor tolerance to chromosomal instability, and facilitate the establishment of an immunologically exhausted tumor microenvironment. Here, we review fundamental aspects of type I IFN signaling and their context-dependent impact on malignant transformation, tumor progression, and response to therapy.
Collapse
Affiliation(s)
- Peter Holicek
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Emma Guilbaud
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Vanessa Klapp
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Strassen, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | | | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York, USA
- Sandra and Edward Meyer Cancer Center, New York, New York, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, New York, USA
| | - Jitka Fucikova
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
2
|
Boivin N, Baillargeon J, Doss PMIA, Roy AP, Rangachari M. Interferon-β suppresses murine Th1 cell function in the absence of antigen-presenting cells. PLoS One 2015; 10:e0124802. [PMID: 25885435 PMCID: PMC4401451 DOI: 10.1371/journal.pone.0124802] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/19/2015] [Indexed: 01/08/2023] Open
Abstract
Interferon (IFN)-β is a front-line therapy for the treatment of the relapsing-remitting form of multiple sclerosis. However, its immunosuppressive mechanism of function remains incompletely understood. While it has been proposed that IFN-β suppresses the function of inflammatory myelin antigen-reactive T cells by promoting the release of immunomodulatory cytokines such as IL-27 from antigen-presenting cells (APCs), its direct effects on inflammatory CD4+ Th1 cells are less clear. Here, we establish that IFN-β inhibits mouse IFN-γ+ Th1 cell function in the absence of APCs. CD4+ T cells express the type I interferon receptor, and IFN-β can suppress Th1 cell proliferation under APC-free stimulation conditions. IFN-β-treated myelin antigen-specific Th1 cells are impaired in their ability to induce severe experimental autoimmune encephalomyelitis (EAE) upon transfer to lymphocyte-deficient Rag1-/- mice. Polarized Th1 cells downregulate IFN-γ and IL-2, and upregulate the negative regulatory receptor Tim-3, when treated with IFN-β in the absence of APCs. Further, IFN-β treatment of Th1 cells upregulates phosphorylation of Stat1, and downregulates phosphorylation of Stat4. Our data indicate that IFN-γ-producing Th1 cells are directly responsive to IFN-β and point to a novel mechanism of IFN-β-mediated T cell suppression that is independent of APC-derived signals.
Collapse
Affiliation(s)
- Nicolas Boivin
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
| | - Joanie Baillargeon
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
| | - Prenitha Mercy Ignatius Arokia Doss
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Graduate Programme in Microbiology and Immunology, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
| | - Andrée-Pascale Roy
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Graduate Programme in Microbiology and Immunology, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
| | - Manu Rangachari
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
- * E-mail:
| |
Collapse
|
3
|
Shen L, Suresh L, Malyavantham K, Kowal P, Xuan J, Lindemann MJ, Ambrus JL. Different Stages of Primary Sjögren’s Syndrome Involving Lymphotoxin and Type 1 IFN. THE JOURNAL OF IMMUNOLOGY 2013; 191:608-13. [DOI: 10.4049/jimmunol.1203440] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
4
|
Hong J, Hutton GJ. Regulatory effects of interferon-β on osteopontin and interleukin-17 expression in multiple sclerosis. J Interferon Cytokine Res 2011; 30:751-7. [PMID: 20874252 DOI: 10.1089/jir.2010.0082] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease characterized by autoimmune inflammation in the central nervous system. Despite over a decade of use of interferon-β (IFN-β) in the treatment of MS, its mechanisms of action are still not fully elucidated. New data now demonstrate that the 2 important proinflammatory cytokines involved in the pathogenesis of MS, osteopontin (OPN) and interleukin-17 (IL-17), are regulated by IFN-β. This review discusses the role of OPN and IL-17 in the development of MS and how the downregulation of the levels of OPN and interleukin-17 contributes to the therapeutic effects of IFN-β in MS.
Collapse
Affiliation(s)
- Jian Hong
- Department of Neurology and Baylor Multiple Sclerosis Center, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | |
Collapse
|
5
|
McFarland AP, Savan R, Wagage S, Addison A, Ramakrishnan K, Karwan M, Duong T, Young HA. Localized delivery of interferon-β by Lactobacillus exacerbates experimental colitis. PLoS One 2011; 6:e16967. [PMID: 21365015 PMCID: PMC3041828 DOI: 10.1371/journal.pone.0016967] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 01/19/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND There have been conflicting reports of the role of Type I interferons (IFN) in inflammatory bowel disease (IBD). Clinical trials have shown potent efficacy of systemic interferon-beta (IFN-β) in inducing remission of ulcerative colitis. Likewise, IFNAR1(-/-) mice display an increased sensitivity to dextran sulfate sodium (DSS)-induced colitis, suggesting Type I IFN play a protective role during inflammation of the gut. Curiously, however, there have also been reports detailing the spontaneous development of IBD in patients receiving systemic IFN-β therapy for multiple sclerosis or hepatitis. METHODOLOGY/PRINCIPAL FINDINGS To investigate the effects of local administration of IFN-β on a murine model of colitis, we developed a transgenic Lactobacillus acidophilus strain that constitutively expresses IFN-β (La-IFN-β). While pretreatment of mice with control Lactobacillus (La-EV) provided slight protective benefits, La-IFN-β increased sensitivity to DSS. Analysis showed colitic mice pretreated with La-IFN-β had increased production of TNF-α, IFN-γ, IL-17A and IL-13 by intestinal tissues and decreased regulatory T cells (Tregs) in their small intestine. Examination of CD103(+) dendritic cells (DCs) in the Peyer's patches revealed that IFNAR1 expression was dramatically reduced by La-IFN-β. Similarly, bone marrow-derived DCs matured with La-IFN-β experienced a 3-fold reduction of IFNAR1 and were impaired in their ability to induce Tregs. CONCLUSIONS/SIGNIFICANCE Our IFNAR1 expression data identifies a correlation between the loss/downregulation of IFNAR1 on DCs and exacerbation of colitis. Our data show that Lactobacillus secreting IFN-β has an immunological effect that in our model results in the exacerbation of colitis. This study underscores that the selection of therapeutics delivered by a bacterial vehicle must take into consideration the simultaneous effects of the vehicle itself.
Collapse
Affiliation(s)
- Adelle P. McFarland
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Ram Savan
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Sagie Wagage
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Augustina Addison
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Karthika Ramakrishnan
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Megan Karwan
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Tri Duong
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Howard A. Young
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| |
Collapse
|
6
|
Abstract
Guillain-Barré syndrome (GBS) is an acute inflammatory demyelinating neuropathy that is associated with long-lasting morbidity and a substantial risk of mortality. The 2 reference treatments, plasma exchange and intravenous immunoglobulins (IVIg), do not change the functional prognosis for the most severely ill patients. The pathogenesis of GBS involves humoral and cellular immune dysfunctions that have only recently been characterised. Antibodies to nerve antigens may participate in complement activation, antibody-dependent macrophage cytotoxicity and reversible conduction failure. The cellular immune reaction is associated with increases in pro-inflammatory cytokines [such as tumour necrosis factor-alpha (TNFalpha)] and matrix metalloproteinases (MMPs; e.g. MMP-9), and a decrease in anti-inflammatory cytokines [such as transforming growth factor-beta1 (TGFbeta1)]. All the changes favour adhesion to and transmigration across the endothelium of immune cells, a key phenomenon associated with GBS. Recovery from GBS is characterised by the normalisation of these changes. Experimental allergic neuritis (EAN), the experimental model of GBS, has strikingly similar immunological characteristics. The usual treatment options for patients with GBS (plasma exchange and IVIg) mainly target the humoral component of the immune response. Interferon-beta (IFNbeta) is a cellular immunomodulator that inhibits antigen presentation and TNFalpha production and binding, and modulates macrophage properties. IFNbeta increases anti-inflammatory T cell functions and the production of anti-inflammatory cytokines, such as TGFbeta1. IFNbeta has important effects on leukodiapedesis, caused by modulating the expression of cell adhesion molecules and the MMP-9 proteinases. It has been used with success in EAN, in some patients with acute exacerbation of chronic inflammatory demyelinating polyneuropathy, and in 1 patient with GBS. The pathophysiology of patients with GBS, an understanding of IFNbeta properties and results of experimental studies support the investigation of IFNbeta in trials of patients with GBS.
Collapse
Affiliation(s)
- A Créange
- Réseau de Neuroimmunologie du Nerf Périphérique (AP/HP), Laboratoire Germen (Inserm E. 0011), Service de Neurologie, Centre Hospitalier Universitaire Henri Mondor, Créteil, France.
| |
Collapse
|
7
|
CpG ODN mediated prevention from ovalbumin-induced anaphylaxis in mouse through B cell pathway. Int Immunopharmacol 2007; 8:351-61. [PMID: 18182251 DOI: 10.1016/j.intimp.2007.10.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Revised: 10/04/2007] [Accepted: 10/23/2007] [Indexed: 11/22/2022]
Abstract
Allergic inflammation is induced by type 2 T helper cell (Th2) and Th2 cytokines such as interleukin (IL)-4, IL-5 and IL-13. These cytokines induce the production of allergen-specific immunoglobulin (Ig)E by B cells, and the ensuing degranulation of mast cells via IgE cross-linking leads to most clinical manifestations of allergic diseases. We examined the ability of immunomodulatory unmethylated CpG oligodeoxynucleotides (ODN), which are potent inducers of Th1 cytokines, to prevent allergic symptoms in mice immunized and sensitized with allergen. Coadministration of CpG ODN with ovalbumin (OVA) before OVA sensitization substantially prevented mice from allergic anaphylaxis representing enhanced circulating concentrations of OVA-specific IgE and histamine, and decreased body temperature. Although CpG ODN provokes an abundance of Th1-skewing cytokines, including IL-12, interferon (IFN)-alpha and IFN-gamma, administration of CpG ODN in IFN-gamma deficient mice inhibited IgE production and prevented from OVA-induced anaphylaxis, indicating a dispensable role of IFN-gamma in mediating these protective effects. In vitro analysis revealed that CpG ODN inhibited class switching from IgM to IgE and IgG1 in response to CD40 and IL-4 in B cells, and this effect did not correlate with up-regulation of IFN-alpha production. These results imply a B cell-intrinsic, T cell-independent mechanism by which CpG ODN directly acts on B cells and inhibits IgE and IgG1 production leading to cause prevention from allergic symptoms.
Collapse
|
8
|
Suppression of autoimmune inflammation of the central nervous system by interleukin 10 secreted by interleukin 27–stimulated T cells. Nat Immunol 2007; 8:1372-9. [DOI: 10.1038/ni1540] [Citation(s) in RCA: 439] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2007] [Accepted: 10/16/2007] [Indexed: 01/10/2023]
|
9
|
Paez A, Rodriguez E, Rendon E, Varela E, Fortoul T, Espinosa B, Masso F, Guevara J, Montaño LF. Altered detection of molecules associated with leukocyte traffic in HUVECs derived from newborns with a strong family history of myocardial infarction. Acta Histochem 2007; 110:42-52. [PMID: 17765294 DOI: 10.1016/j.acthis.2007.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Revised: 05/17/2007] [Accepted: 05/22/2007] [Indexed: 01/21/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease. As such, recruitment of immune cells is a significant event. Tightly controlled signaling molecules regulate leukocyte adhesion and migration to the tissues. The aim of this study was to determine if human umbilical vein endothelial cells (HUVECs) derived from healthy newborns with a strong family history of myocardial infarction (FHMI) showed variations in the presence of molecules related with leukocyte traffic and migration, in comparison to control healthy newborns. For this purpose, we evaluated the labeling of sialic acid containing glycoproteins, tight junction claudins and the cytoskeleton, using lectin- and immunocytochemistry in HUVECs from individuals with and without a strong FHMI. Our results show important differences in the labeling of alpha-2,3 or alpha-2,6 sialic acid-containing glycoconjugates, a disarrangement of actin filaments secondary to the absence of cytoplasmic claudin-5 immunopositivity and an increase in the binding of FHMI HUVECs to CD3+ Jurkat cells. It is possible that these differences relate to a predisposition for early appearance of atherosclerotic lesions.
Collapse
Affiliation(s)
- Araceli Paez
- Departamento Biología Celular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Col. Seccion 16, México 14080, México
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Byrnes AA, Li DY, Park K, Thompson D, Mocilnikar C, Mohan P, Molleston JP, Narkewicz M, Zhou H, Wolf SF, Schwarz KB, Karp CL. Modulation of the IL-12/IFN-γ axis by IFN-α therapy for hepatitis C. J Leukoc Biol 2006; 81:825-34. [PMID: 17148690 DOI: 10.1189/jlb.1006622] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although IFN-alpha forms the foundation of therapy for chronic hepatitis C, only a minority of patients has a sustained response to IFN-alpha alone. The antiviral activities of IFN-alpha formed the rationale for its use in viral hepatitis. However, IFN-alpha and the other Type I IFNs are also pleiotropic immune regulators. Type I IFNs can promote IFN-gamma production by activating STAT4 but can also inhibit production of IL-12, a potent activator of STAT4 and IFN-gamma production. The efficacy of IFN-alpha in the treatment of hepatitis C may therefore depend in part on the balance of IFN-gamma-inducing and IL-12-suppressing effects. We characterized the effects of pegylated IFN-alpha therapy for hepatitis C on the capacity of patients' PBMC to produce IL-12 and IFN-gamma ex vivo. Cells from patients with a sustained virological response to therapy had significantly greater levels of IFN-alpha-driven IFN-gamma production prior to treatment than those from nonresponding patients. No differences in pretreatment IL-12 productive capacity were seen between patient groups. However, therapy with IFN-alpha led to suppression of inducible IL-12 production throughout the course of therapy in both groups of patients.
Collapse
Affiliation(s)
- Adriana A Byrnes
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Teige I, Liu Y, Issazadeh-Navikas S. IFN-beta inhibits T cell activation capacity of central nervous system APCs. THE JOURNAL OF IMMUNOLOGY 2006; 177:3542-53. [PMID: 16951313 DOI: 10.4049/jimmunol.177.6.3542] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have previously investigated the physiological effects of IFN-beta on chronic CNS inflammation and shown that IFN-beta(-/-) mice develop a more severe experimental autoimmune encephalomyelitis than their IFN-beta(+/-) littermates. This result was shown to be associated with a higher activation state of the glial cells and a higher T cell cytokine production in the CNS. Because this state suggested a down-regulatory effect of IFN-beta on CNS-specific APCs, these results were investigated further. We report that IFN-beta pretreatment of astrocytes and microglia (glial cells) indeed down-modulate their capacity to activate autoreactive Th1 cells. First, we investigated the intrinsic ability of glial cells as APCs and report that glial cells prevent autoreactive Th1 cells expansion while maintaining Ag-specific T cell effector functions. However, when the glial cells are treated with IFN-beta before coculture with T cells, the effector functions of T cells are impaired as IFN-gamma, TNF-alpha, and NO productions are decreased. Induction of the T cell activation marker, CD25 is also reduced. This suppression of T cell response is cell-cell dependent, but it is not dependent on a decrease in glial expression of MHC class II or costimulatory molecules. We propose that IFN-beta might exert its beneficial effects mainly by reducing the Ag-presenting capacity of CNS-specific APCs, which in turn inhibits the effector functions of encephalitogenic T cells. This affect is of importance because activation of encephalitogenic T cells within the CNS is a prerequisite for the development of a chronic progressive CNS inflammation.
Collapse
Affiliation(s)
- Ingrid Teige
- Neuroinflammation Unit, Section for Immunology, Institute for Experimental Medical Science, Lund University, Lund, Sweden
| | | | | |
Collapse
|
12
|
Affiliation(s)
- Stergios Moschos
- University of Pittsburgh Cancer Institute Melanoma and Skin Cancer Program, Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh, School of Medicine, PA, USA
| | | | | |
Collapse
|
13
|
Nagai T, Devergne O, Mueller TF, Perkins DL, van Seventer JM, van Seventer GA. Timing of IFN-beta exposure during human dendritic cell maturation and naive Th cell stimulation has contrasting effects on Th1 subset generation: a role for IFN-beta-mediated regulation of IL-12 family cytokines and IL-18 in naive Th cell differentiation. THE JOURNAL OF IMMUNOLOGY 2004; 171:5233-43. [PMID: 14607924 DOI: 10.4049/jimmunol.171.10.5233] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Type I IFNs, IFN-alpha and IFN-beta, are early effectors of innate immune responses against microbes that can also regulate subsequent adaptive immunity by promoting antimicrobial Th1-type responses. In contrast, the ability of IFN-beta to inhibit autoimmune Th1 responses is thought to account for some of the beneficial effects of IFN-beta therapy in the treatment of relapsing remitting multiple sclerosis. To understand the basis of the paradoxical effects of IFN-beta on the expression of Th1-type immune responses, we developed an in vitro model of monocyte-derived dendritic cell (DC)-dependent, human naive Th cell differentiation, in which one can observe both positive and negative effects of IFN-beta on the generation of Th1 cells. In this model we found that the timing of IFN-beta exposure determines whether IFN-beta will have a positive or a negative effect on naive Th cell differentiation into Th1 cells. Specifically, the presence of IFN-beta during TNF-alpha-induced DC maturation strongly augments the capacity of DC to promote the generation of IFN-gamma-secreting Th1 cells. In contrast, exposure to IFN-beta during mature DC-mediated primary stimulation of naive Th cells has the opposite effect, in that it inhibits Th1 cell polarization and promotes the generation of an IL-10-secreting T cell subset. Studies with blocking mAbs and recombinant cytokines indicate that the mechanism by which IFN-beta mediates these contrasting effects on Th1 cell generation is at least in part by differentially regulating DC expression of IL-12 family cytokines (IL-12 and/or IL-23, and IL-27) and IL-18.
Collapse
Affiliation(s)
- Taro Nagai
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA
| | | | | | | | | | | |
Collapse
|
14
|
Trobonjaca Z, Kröger A, Stober D, Leithäuser F, Möller P, Hauser H, Schirmbeck R, Reimann J. Activating immunity in the liver. II. IFN-beta attenuates NK cell-dependent liver injury triggered by liver NKT cell activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3763-70. [PMID: 11937527 DOI: 10.4049/jimmunol.168.8.3763] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Dendritic cell (DC)-dependent activation of liver NKT cells triggered by a single i.v. injection of a low dose (10-100 ng/mouse) of alpha-galactosyl ceramide (alphaGalCer) into mice induces liver injury. This response is particularly evident in HBs-tg B6 mice that express a transgene-encoded hepatitis B surface Ag in the liver. Liver injury following alphaGalCer injection is suppressed in mice depleted of NK cells, indicating that NK cells play a role in NK T cell-initiated liver injury. In vitro, liver NKT cells provide a CD80/86-dependent signal to alphaGalCer-pulsed liver DC to release IL-12 p70 that stimulates the IFN-gamma response of NKT and NK cells. Adoptive transfer of NKT cell-activated liver DC into the liver of nontreated, normal (immunocompetent), or immunodeficient (RAG(-/-) or HBs-tg/RAG(-/-)) hosts via the portal vein elicited IFN-gamma responses of liver NK cells in situ. IFN-beta down-regulates the pathogenic IL-12/IFN-gamma cytokine cascade triggered by NKT cell/DC/NK cell interactions in the liver. Pretreating liver DC in vitro with IFN-beta suppressed their IL-12 (but not IL-10) release in response to CD40 ligation or specific (alphaGalCer-dependent) interaction with liver NKT cells and down-regulated the IFN-gamma response of the specifically activated liver NKT cells. In vivo, IFN-beta attenuated the NKT cell-triggered induction of liver immunopathology. This study identifies interacting subsets of the hepatic innate immune system (and cytokines that up- and down-regulate these interactions) activated early in immune-mediated liver pathology.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, CD/physiology
- B7-1 Antigen/physiology
- B7-2 Antigen
- Cells, Cultured
- Coculture Techniques
- Cytotoxicity, Immunologic/genetics
- Cytotoxicity, Immunologic/immunology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/transplantation
- Galactosylceramides/administration & dosage
- Galactosylceramides/pharmacology
- Immunosuppressive Agents/pharmacology
- Injections, Intravenous
- Interferon-beta/pharmacology
- Interleukin-12/antagonists & inhibitors
- Interleukin-12/metabolism
- Interleukin-12/physiology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Liver/immunology
- Liver/pathology
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Zlatko Trobonjaca
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Salio M, Shepherd D, Dunbar PR, Palmowski M, Murphy K, Wu L, Cerundolo V. Mature dendritic cells prime functionally superior melan-A-specific CD8+ lymphocytes as compared with nonprofessional APC. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:1188-97. [PMID: 11466333 DOI: 10.4049/jimmunol.167.3.1188] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Priming of melan-A(26/27-35)-specific CTL occurs only in a fraction of late stage melanoma patients, whereas during the early stages of the disease and in healthy volunteers, melan-A CTL have functional and phenotypic markers consistent with a naive phenotype. To study the requirements for expansion of naive melan-A CTL from healthy donors, we set up an in vitro priming protocol and, using tetramer assays, we demonstrate that the activity and phenotype of the expanded melan-A CTL are profoundly influenced by the type of APC used. Priming by nonprofessional APC leads to expansion of melan-A CTL with reduced cytolytic activity and low level of IFN-gamma secretion. In contrast, mature dendritic cells (DC) expand cytolytic and IFN-gamma-producing melan-A CTL. Priming by mature DC is also efficient at low peptide concentration and requires only one round of stimulation. Finally, we observed that a significant fraction of CD45RO(+) melan-A CTL primed by mature DC expresses high levels of the homing receptor CD62L, whereas CTL primed by nonprofessional APC express CD62L in lower percentages and at lower levels. These results suggest that suboptimal priming by nonprofessional APC could account for the presence in vivo of dysfunctional cells and strongly support the immunotherapeutic use of mature DC for expansion of effector and memory Ag-specific CTL.
Collapse
Affiliation(s)
- M Salio
- Molecular Immunology Group, Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, OX3 9DS, U.K
| | | | | | | | | | | | | |
Collapse
|
16
|
Szabo G, Catalano D, Bellerose G, Mandrekar P. Interferon alpha and Alcohol Augment Nuclear Regulatory Factor-kappaB Activation in HepG2 Cells, and Interferon alpha Increases Pro-Inflammatory Cytokine Production. Alcohol Clin Exp Res 2001. [DOI: 10.1111/j.1530-0277.2001.tb02335.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
17
|
Interferon ?? and Alcohol Augment Nuclear Regulatory Factor-??B Activation in HepG2 Cells, and Interferon ?? Increases Pro-Inflammatory Cytokine Production. Alcohol Clin Exp Res 2001. [DOI: 10.1097/00000374-200108000-00015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
18
|
Jungo F, Dayer JM, Modoux C, Hyka N, Burger D. IFN-beta inhibits the ability of T lymphocytes to induce TNF-alpha and IL-1beta production in monocytes upon direct cell-cell contact. Cytokine 2001; 14:272-82. [PMID: 11444907 DOI: 10.1006/cyto.2001.0884] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumour necrosis factor (TNF)-alpha and interleukin (IL-)1beta, essential players in the pathogenesis of immuno-inflammatory diseases, are strongly induced in monocytes by direct contact with stimulated T lymphocytes. The present study shows that the latter mechanism is inhibited by interferon (IFN)-beta. In co-cultures of autologous T lymphocytes and monocytes stimulated by phytohaemagglutinin (PHA), IFN-beta inhibited the production of TNF-alpha and IL-1beta by 88 and 98%, respectively, whereas the simultaneous production of IL-1 receptor antagonist (IL-1Ra), was enhanced two-fold. The latter effects of IFN-beta were independent of modulations in IFN-gamma, IL-4 and IL-10 production. When monocytes were activated by plasma membranes of stimulated T cells, IFN-beta slightly inhibited the production of TNF-alpha and IL-1beta, while enhancing 1.5-fold that of IL-1Ra. The latter effect correlated with the persistence of high steady-state levels of IL-1Ra mRNA after 24 h of activation. Membranes isolated from T lymphocytes that had been stimulated in the presence of IFN-beta displayed a 80% decrease in their capacity to induce the production of IL-1beta and TNF-alpha in monocytes, whereas IL-1Ra induction was decreased by only 32%. These results demonstrate that IFN-beta modulates contact-mediated activation of monocytes by acting on both T lymphocytes and monocytes, decreasing the ability of T lymphocytes to induce TNF-alpha and IL-1beta production in monocytes and directly enhancing the production of IL-1Ra in the latter cells.
Collapse
Affiliation(s)
- F Jungo
- Division of Immunology and Allergy, Clinical Immunology Unit (Hans Wilsdorf Laboratory), Department of Internal Medicine, University Hospital, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
19
|
Karp CL, van Boxel-Dezaire AH, Byrnes AA, Nagelkerken L. Interferon-β in multiple sclerosis: altering the balance of interleukin-12 and interleukin-10? Curr Opin Neurol 2001; 14:361-8. [PMID: 11371761 DOI: 10.1097/00019052-200106000-00016] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Interferon-beta is a remarkably pleiotropic molecule. Antiviral, pro- and antiproliferative, pro- and antiapoptotic, and complex immunoregulatory activities have all been described. The precise mechanism(s) that underlie the beneficial effects of interferon-beta in multiple sclerosis remain poorly understood; this has hindered progress in the search for more effective therapies. An increasing body of literature supports the hypothesis that interferon-beta-mediated changes in the production and activities of the immunoregulatory cytokines interleukin-12 and interleukin-10 are important to the therapeutic benefits of interferon-beta in multiple sclerosis. These data are reviewed here.
Collapse
Affiliation(s)
- C L Karp
- Molecular Immunology Section, Children's Hospital Research Foundation, University of Cincinnati, Cincinnati, Ohio, USA.
| | | | | | | |
Collapse
|
20
|
Tuohy VK, Yu M, Yin L, Mathisen PM, Johnson JM, Kawczak JA. Modulation of the IL-10/IL-12 cytokine circuit by interferon-beta inhibits the development of epitope spreading and disease progression in murine autoimmune encephalomyelitis. J Neuroimmunol 2000; 111:55-63. [PMID: 11063821 DOI: 10.1016/s0165-5728(00)00384-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
IFN-beta has been shown to be effective in the treatment of multiple sclerosis (MS). However, the primary mechanism by which IFN-beta mediates its therapeutic effect remains unclear. Recent studies indicate that under defined conditions, IFN-beta may downregulate DC expression of IL-12. We and others have shown that IFN-beta may also downregulate IL-10. In light of the recently proposed paradigm that an IL-10/IL-12 immunoregulatory circuit controls susceptibility to autoimmune disease, we examined the effect of IFN-beta on the development and behavior of the autoreactive T cell repertoire during experimental autoimmune encephalomyelitis (EAE), an animal model sharing many features with MS. SWXJ mice were immunized with the immunodominant p139-151 determinant of myelin proteolipid protein (PLP), and at onset of EAE were treated every other day with IFN-beta. After eight weeks of treatment, we assessed autoreactivity and observed no significant IFN-beta effect on splenocyte proliferation or splenocyte production of IFN-gamma, IL-2, IL-4, or IL-5 in response to the priming determinant used to initiate disease. However, in IFN-beta treated mice, the cytokine profile in response to the priming immunogen was significantly skewed toward an increased production of IL-10 and a concurrent decreased production of IL-12. Moreover, the in vivo modulation of the IL-10/IL-12 immunoregulatory circuit in response to the priming immunogen was accompanied by an aborted development of epitope spreading. Our results indicate that IFN-beta induces a reciprocal modulation of the IL-10/IL-12 cytokine circuit in vivo. This skewed autoreactivity establishes an inflammatory microenvironment that effectively prevents endogenous self-priming thereby inhibiting the progression of disease associated with epitope spreading.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Animals
- Disease Progression
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Female
- Immunization
- Interferon-beta/pharmacology
- Interleukin-10/immunology
- Interleukin-10/metabolism
- Interleukin-12/immunology
- Interleukin-12/metabolism
- Mice
- Mice, Inbred Strains
- Myelin Proteolipid Protein/immunology
- Myelin Proteolipid Protein/pharmacology
- Peptide Fragments/immunology
- Peptide Fragments/pharmacology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th2 Cells/drug effects
- Th2 Cells/immunology
Collapse
Affiliation(s)
- V K Tuohy
- Department of Immunology, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, 44195, Cleveland, OH, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Rook GA, Ristori G, Salvetti M, Giovannoni G, Thompson EJ, Stanford JL. Bacterial vaccines for the treatment of multiple sclerosis and other autoimmune disorders. IMMUNOLOGY TODAY 2000; 21:503-8. [PMID: 11071529 DOI: 10.1016/s0167-5699(00)01700-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- G A Rook
- Dept of Medical Microbiology, Windeyer Institute of Medical Sciences, Royal Free and University College Medical School, London, UK.
| | | | | | | | | | | |
Collapse
|
22
|
Cremer I, Vieillard V, Sautès-Fridman C, De Maeyer E. Inhibition of human immunodeficiency virus transmission to CD4+ T cells after gene transfer of constitutively expressed interferon beta to dendritic cells. Hum Gene Ther 2000; 11:1695-703. [PMID: 10954903 DOI: 10.1089/10430340050111340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CD34(+)-derived dendritic cells (DCs) can be infected by the T cell-tropic HIVLAI strain, but are poorly permissive for efficient virus production. However, HIVLAI-infected DCs are able to transmit a vigorous cytopathic infection to activated CD4(+) T cells. We show that DCs differentiated from CD34(+) cells can be efficiently transduced by a retroviral vector carrying the IFN-beta coding sequence. This results in resistance to infection by HIV as shown by a threefold reduction in the HIV DNA copy number per cell, and by inhibition of HIV transmission from DCs to CD4(+) T cells. Moreover, constitutive IFN-beta production by DCs increases the synthesis of IL-12 and IFN-gamma Th1-type cytokines and of the beta-chemokines MIP-1alpha, MIP-1beta, and RANTES. This indicates that IFN-beta transduction of DCs blocks HIV infection and viral transmission to CD4(+) T cells, and could favor cellular immune responses in HIV-infected patients.
Collapse
Affiliation(s)
- I Cremer
- Laboratorie d'Immunologie Cellulaire et Clinique, INSERM U255, Institut Curie, Paris, France.
| | | | | | | |
Collapse
|
23
|
Interferon-α and -β inhibit the in vitro differentiation of immunocompetent human dendritic cells from CD14+ precursors. Blood 2000. [DOI: 10.1182/blood.v96.1.210.013k52_210_217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dendritic cell (DC) precursors and immature DC reside in epithelium where they encounter pathogens and cytokines, which stimulate their differentiation. We hypothesized that type-I interferons (IFN- and -β), cytokines that are produced early in the innate immune response against viruses and some bacteria, may influence DC differentiation and function. To examine this possibility, we used an in vitro model of DC differentiation in which initial culture of human CD14+monocytes with granulocyte–macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-4 generates immature DC, and subsequent culture with tumor necrosis factor (TNF)- drives the final development into mature DC. We found in this model that IFN-/β, added from the initiation of the culture on, significantly reduced the survival and altered the morphology and differentiation of DC. TNF-–dependent maturation of IFN-β–treated immature DC led to cells with reduced expression of CD1a, CD40, CD54, and CD80 when compared with mature DC controls. IFN-/β–treated DC further had a reduced capacity to induce naive Th-cell proliferation through allostimulation or anti-CD3 monoclonal antibody stimulation. In addition, IFN-/β–treated DC secreted less IL-12 upon stimulation with Staphylococcus aureus Cowan strain or with CD4+ T cells, and this decrease correlated directly with their inability to support CD4+ T-cell secretion of IFN-γ, even though T-cell lymphotoxin production was unaffected. These findings indicate that type-I IFNs can influence the generation of acquired immune responses by modifying T-helper cell differentiation through the regulation of DC differentiation and function.
Collapse
|
24
|
Interferon-α and -β inhibit the in vitro differentiation of immunocompetent human dendritic cells from CD14+ precursors. Blood 2000. [DOI: 10.1182/blood.v96.1.210] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Dendritic cell (DC) precursors and immature DC reside in epithelium where they encounter pathogens and cytokines, which stimulate their differentiation. We hypothesized that type-I interferons (IFN- and -β), cytokines that are produced early in the innate immune response against viruses and some bacteria, may influence DC differentiation and function. To examine this possibility, we used an in vitro model of DC differentiation in which initial culture of human CD14+monocytes with granulocyte–macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-4 generates immature DC, and subsequent culture with tumor necrosis factor (TNF)- drives the final development into mature DC. We found in this model that IFN-/β, added from the initiation of the culture on, significantly reduced the survival and altered the morphology and differentiation of DC. TNF-–dependent maturation of IFN-β–treated immature DC led to cells with reduced expression of CD1a, CD40, CD54, and CD80 when compared with mature DC controls. IFN-/β–treated DC further had a reduced capacity to induce naive Th-cell proliferation through allostimulation or anti-CD3 monoclonal antibody stimulation. In addition, IFN-/β–treated DC secreted less IL-12 upon stimulation with Staphylococcus aureus Cowan strain or with CD4+ T cells, and this decrease correlated directly with their inability to support CD4+ T-cell secretion of IFN-γ, even though T-cell lymphotoxin production was unaffected. These findings indicate that type-I IFNs can influence the generation of acquired immune responses by modifying T-helper cell differentiation through the regulation of DC differentiation and function.
Collapse
|
25
|
Price GE, Gaszewska-Mastarlarz A, Moskophidis D. The role of alpha/beta and gamma interferons in development of immunity to influenza A virus in mice. J Virol 2000; 74:3996-4003. [PMID: 10756011 PMCID: PMC111913 DOI: 10.1128/jvi.74.9.3996-4003.2000] [Citation(s) in RCA: 153] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/1999] [Accepted: 01/29/2000] [Indexed: 01/12/2023] Open
Abstract
During influenza virus infection innate and adaptive immune defenses are activated to eliminate the virus and thereby bring about recovery from illness. Both arms of the adaptive immune system, antibody neutralization of free virus and termination of intracellular virus replication by antiviral cytotoxic T cells (CTLs), play pivotal roles in virus elimination and protection from disease. Innate cytokine responses, such as alpha/beta interferon (IFN-alpha/beta) or IFN-gamma, can have roles in determining the rate of virus replication in the initial stages of infection and in shaping the initial inflammatory and downstream adaptive immune responses. The effect of these cytokines on the replication of pneumotropic influenza A virus in the respiratory tract and in the regulation of adaptive antiviral immunity was examined after intranasal infection of mice with null mutations in receptors for IFN-alpha/beta, IFN-gamma, and both IFNs. Virus titers in the lungs of mice unable to respond to IFNs were not significantly different from congenic controls for both primary and secondary infection. Likewise the mice were comparably susceptible to X31 (H3N2) influenza virus infection. No significant disruption to the development of normal antiviral CTL or antibody responses was observed. In contrast, mice bearing the disrupted IFN-alpha/beta receptor exhibited accelerated kinetics and significantly higher levels of neutralizing antibody activity during primary or secondary heterosubtypic influenza virus infection. Thus, these observations reveal no significant contribution for IFN-controlled pathways in shaping acute or memory T-cell responses to pneumotropic influenza virus infection but do indicate some role for IFN-alpha/beta in the regulation of antibody responses. Recognizing the pivotal role of CTLs and antibody in virus clearance, it is reasonable to assume a redundancy in IFN-mediated antiviral effects in pulmonary influenza. However, IFN-alpha/beta seems to be a valid factor in determining tissue tropism and replicative rates of highly virulent influenza virus strains as reported previously by others, and this aspect is discussed here.
Collapse
Affiliation(s)
- G E Price
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia 30912-3175, USA
| | | | | |
Collapse
|
26
|
Pacocha SE, Oriente A, Lichtenstein LM, Huang SK, Essayan DM. Interferon-beta induces selective enhancement of antigen-specific T cell responses. J Interferon Cytokine Res 2000; 20:383-9. [PMID: 10805373 DOI: 10.1089/107999000312324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Interferon-beta (IFN-beta) inhibits mitogen-induced T cell responses, in part through downregulation of interleukin-12 (IL-12) or upregulation of IL-10. We have reexamined these findings using ragweed (RW) stimulated or tetanus toxoid (TT)-stimulated human peripheral blood mononuclear cells (PBMC) and nontransformed, antigen-specific, human Th0, Th1, and Th2 clones. IFN-beta induced concentration-dependent inhibition of phytohemagglutinin (PHA)-stimulated PBMC proliferation and enhancement of RW-stimulated or TTstimulated PBMC proliferation. Monocyte depletion of PBMC isolates resulted in concentration-dependent inhibition of RW-driven or TT-driven proliferation by IFN-beta. This response was unaltered by the addition of either exogenous recombinant human IL-12 (rHuIL-12) or saturating concentrations of anti-IL-10. Moreover, addition of exogenous rHuIL-10 to nondepleted RW-driven or TT-driven PBMC cultures did not alter the concentration-dependent enhancement of antigen-driven proliferation induced by IFN-beta. Th0, Th1, and Th2 clones stimulated in the presence of antigen and autologous, irradiated PBMC displayed concentration-dependent inhibition of proliferation in the presence of IFN-beta that was unaltered by the addition of either exogenous rHuIL-12 or a saturating concentration of anti-IL-10. Finally, whereas IFN-beta inhibited antigen-driven generation of IL-5, IL-12, IL-13, and IFN-gamma, IFN-beta enhanced generation of both IL-4 and IL-10. Thus, IFN-beta, induces a selective, IL-10-independent and IL-12-independent upregulation of antigen-specific T cell responses, supporting the role of IFN-beta as an immunomodulatory rather than an antiproliferative/immunosuppressive cytokine.
Collapse
Affiliation(s)
- S E Pacocha
- Johns Hopkins University School of Medicine, Department of Internal Medicine, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
27
|
Cremer I, Vieillard V, De Maeyer E. Retrovirally mediated IFN-beta transduction of macrophages induces resistance to HIV, correlated with up-regulation of RANTES production and down-regulation of C-C chemokine receptor-5 expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:1582-7. [PMID: 10640778 DOI: 10.4049/jimmunol.164.3.1582] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Constitutive expression of IFN-beta by HIV target cells may be an alternative or complementary therapeutic approach for the treatment of AIDS. We show that macrophages derived from CD34+ cells from umbilical cord blood can be efficiently transduced by a retroviral vector carrying the IFN-beta coding sequence. This results in resistance to infection by a macrophage-tropic HIV type 1, as shown by the drastic reduction in the HIV DNA copy number per cell and in p24 release. Moreover, IFN-beta transduction totally blocked secretion of proinflammatory cytokines after HIV infection. The constitutive IFN-beta production also resulted in an increased production of IL-12 and IFN-gamma Th1-type cytokines and of the beta-chemokines macrophage-inflammatory protein-1alpha, macrophage-inflammatory protein-1beta, and RANTES. RANTES was found to be involved in the HIV resistance observed, and this was correlated with a down-regulation of the CCR-5 HIV entry coreceptor. These results demonstrate the feasibility and the efficacy of such IFN-beta-mediated gene therapy. In addition to inhibiting HIV replication, IFN-beta transduction could have beneficial immune effects in HIV-infected patients by favoring cellular immune responses.
Collapse
Affiliation(s)
- I Cremer
- Equipe de l'Interferon et des Cytokines, Unité Mixte de Recherche 146, Centre National de la Recherche Scientifique Institut Curie, Orsay, France.
| | | | | |
Collapse
|
28
|
McRae BL, Beilfuss BA, van Seventer GA. IFN-beta differentially regulates CD40-induced cytokine secretion by human dendritic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:23-8. [PMID: 10604988 DOI: 10.4049/jimmunol.164.1.23] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have previously shown that IFN-beta, a key cytokine associated with the early phase of the innate host defense, can prevent the generation of human Th1 cells. Specifically, we demonstrated that IFN-beta prevents the in vitro monocyte-derived mature dendritic cell (DC)-dependent differentiation of naive Th cells into IFN-gamma-secreting Th cells, as a result of its ability to inhibit DC IL-12 secretion. The goal of the present study was to identify how IFN-beta negatively regulates IL-12 secretion by DC. We report that in our Th cell differentiation model, DC IL-12 secretion is dependent on the CD40L/CD40 accessory pathway, and, utilizing a Th cell-free system, we find that IFN-beta inhibits anti-CD40 mAb-induced DC secretion of the p40 chain of the IL-12 heterodimer. In addition, we show that IFN-beta-mediated inhibition of CD40 signaling does not interfere with all signaling pathways emanating from CD40, since anti-CD40 mAb-induced DC IL-6 secretion is augmented by IFN-beta. Thus, our results demonstrate that signaling from CD40 is differentially regulated by IFN-beta. A second critical element of innate immunity involves the response against components of bacterial membranes such as LPS. DC respond to LPS by secreting IL-6 and IL-12. In contrast to CD40-dependent IL-6 and IL-12 secretion, we find that LPS-induced DC secretion of p40 IL-12 and IL-6 is not affected by IFN-beta. Our findings show that IFN-beta influences the generation of acquired immune responses through its regulation of CD40-dependent DC functions.
Collapse
Affiliation(s)
- B L McRae
- Committee on Immunology, Department of Pathology, Division of Biological Sciences, University of Chicago, IL 60637, USA
| | | | | |
Collapse
|
29
|
Karp CL, Biron CA, Irani DN. Interferon beta in multiple sclerosis: is IL-12 suppression the key? IMMUNOLOGY TODAY 2000; 21:24-8. [PMID: 10637555 DOI: 10.1016/s0167-5699(99)01541-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- C L Karp
- Depts of Medicine and Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
30
|
Luca ME, Visser L, Lucas CJ, Nagelkerken L. IFN-beta modulates specific T cell responses in vitro but does not affect Experimental Autoimmune Encephalomyelitis in the SJL mouse. J Neuroimmunol 1999; 100:190-6. [PMID: 10695729 DOI: 10.1016/s0165-5728(99)00198-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, mouse recombinant IFN-beta was shown to favor PLP139-151-specific Th2 responses in vitro, by inhibiting IFN-gamma production and stimulating IL-4 and IL-10 production. IFN-beta (5000 U/day) failed to prevent the development or severity of EAE induced with PLP139-151. Whereas efficacy of IL-10 was found in the B. pertussis assisted but not in the pertussigen-assisted EAE model, both models appeared insensitive to IFN-beta. Also the combination of (suboptimal) IL-10 and IFN-beta appeared ineffective in inhibiting disease. However, the PLP139-151-specific IL-10 production by T cells from these mice appeared significantly more sensitive to the stimulatory effect of IFN-beta in vitro. It is concluded that despite its Th2 promoting effects, IFN-beta is not effective in inhibiting EAE in this study.
Collapse
Affiliation(s)
- M E Luca
- Division of Immunological and Infectious Diseases, TNO Prevention and Health, Leiden, Netherlands.
| | | | | | | |
Collapse
|
31
|
Upregulation of Integrin 6/β1 and Chemokine Receptor CCR1 by Interleukin-12 Promotes the Migration of Human Type 1 Helper T Cells. Blood 1999. [DOI: 10.1182/blood.v94.9.2981.421k27_2981_2989] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD4+ T helper 1 (Th1) cells and Th2 cells are distinguished based on the pattern of cytokines they are able to produce. Selectin ligands and chemokine receptors are differentially expressed in Th1 and Th2 cells, providing a basis for tissue-specific recruitment of helper T-cell subsets. However, the modes and mechanisms regulating tissue-specific localization of Th1 and Th2 cells are still largely unknown. Here, we show the preferential expression on Th1 cells of the integrin 6/β1, which is distinctly regulated by the Th1-inducing cytokines interleukin-12 (IL-12) and interferon-alfa (IFN-). The pattern of integrin 6/β1 regulation closely mirrors that of the chemokine receptor CCR1. Analysis of signal transducer and activator of transcription 4 (Stat4) activation by IL-12 and IFN- shows distinct signaling kinetics by these cytokines, correlating with the pattern of CCR1 and integrin 6/β1 expression. Unlike IFN-, the ability of IL-12 to generate prolonged intracellular signals appears to be critical for inducing integrin 6/β1 upregulation in Th1 cells. The expression and upregulation of CCR1 and 6/β1 integrin promotes the migration of Th1 cells. These findings suggest that the exquisite regulation of integrin 6/β1 and CCR1 may play an important role in tissue-specific localization of Th1 cells.
Collapse
|
32
|
Lock C, Oksenberg J, Steinman L. The role of TNFalpha and lymphotoxin in demyelinating disease. Ann Rheum Dis 1999; 58 Suppl 1:I121-8. [PMID: 10577988 PMCID: PMC1766588 DOI: 10.1136/ard.58.2008.i121] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- C Lock
- Department of Neurology and Neurological Sciences, Stanford University, Beckman Center B002, Stanford, CA 94305, USA
| | | | | |
Collapse
|
33
|
Upregulation of Integrin 6/β1 and Chemokine Receptor CCR1 by Interleukin-12 Promotes the Migration of Human Type 1 Helper T Cells. Blood 1999. [DOI: 10.1182/blood.v94.9.2981] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
CD4+ T helper 1 (Th1) cells and Th2 cells are distinguished based on the pattern of cytokines they are able to produce. Selectin ligands and chemokine receptors are differentially expressed in Th1 and Th2 cells, providing a basis for tissue-specific recruitment of helper T-cell subsets. However, the modes and mechanisms regulating tissue-specific localization of Th1 and Th2 cells are still largely unknown. Here, we show the preferential expression on Th1 cells of the integrin 6/β1, which is distinctly regulated by the Th1-inducing cytokines interleukin-12 (IL-12) and interferon-alfa (IFN-). The pattern of integrin 6/β1 regulation closely mirrors that of the chemokine receptor CCR1. Analysis of signal transducer and activator of transcription 4 (Stat4) activation by IL-12 and IFN- shows distinct signaling kinetics by these cytokines, correlating with the pattern of CCR1 and integrin 6/β1 expression. Unlike IFN-, the ability of IL-12 to generate prolonged intracellular signals appears to be critical for inducing integrin 6/β1 upregulation in Th1 cells. The expression and upregulation of CCR1 and 6/β1 integrin promotes the migration of Th1 cells. These findings suggest that the exquisite regulation of integrin 6/β1 and CCR1 may play an important role in tissue-specific localization of Th1 cells.
Collapse
|
34
|
Wely CAV, Beverley PCL, Brett SJ, Britten CJ, Tite JP. Expression of L-Selectin on Th1 Cells Is Regulated by IL-12. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.3.1214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
L-selectin has become established as a key molecule in the recirculation of naïve T cells from the blood to peripheral lymph nodes, yet little is known about its role in the migration of effector or memory cells. While differentiating naïve CD4+ T cells into Th1 and Th2 subsets in vitro, it was noted that L-selectin levels were maintained on the Th1 subset of cells. The expression of L-selectin on the Th1 cells appeared to be dependent on the presence of IL-12. Th2 cells, differentiated in the absence of IL-12, failed to maintain L-selectin expression. Coculture with IL-12, IL-18, IL-4, TNF-α, or IFN-α, -β, or -γ demonstrated a dependence on IL-12 alone for L-selectin expression. In addition, the inclusion of heat-killed Listeria monocytogenes in the cultures also maintained L-selectin expression on the Th1 cells. In all cultures, the maintenance of L-selectin on the T cell surface could be blocked by the inclusion of anti-IL-12 Abs. Analysis of the mRNA levels for L-selectin in T cells, differentiated in the presence or absence of IL-12, showed that the cytokine appears to exert its effect on L-selectin at the transcriptional level. Given the key role played by IL-12 in the differentiation of naïve T cells into the Th1 subset, the observation that IL-12 can also regulate L-selectin expression has implications for the migration of Th1 effector cells both through the lymphatic system and to sites of inflammation.
Collapse
Affiliation(s)
- Cathy A. van Wely
- *Immunology Unit, Glaxo Wellcome Medicines Research Centre, Stevenage, United Kingdom; and
| | - Peter C. L. Beverley
- †Edward Jenner Institute for Vaccine Research, Compton, Berkshire, United Kingdom
| | - Sara J. Brett
- *Immunology Unit, Glaxo Wellcome Medicines Research Centre, Stevenage, United Kingdom; and
| | - Christopher J. Britten
- *Immunology Unit, Glaxo Wellcome Medicines Research Centre, Stevenage, United Kingdom; and
| | - John P. Tite
- *Immunology Unit, Glaxo Wellcome Medicines Research Centre, Stevenage, United Kingdom; and
| |
Collapse
|
35
|
Rozera C, Carlei D, Lollini PL, De Giovanni C, Musiani P, Di Carlo E, Belardelli F, Ferrantini M. Interferon (IFN)-beta gene transfer into TS/A adenocarcinoma cells and comparison with IFN-alpha: differential effects on tumorigenicity and host response. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 154:1211-22. [PMID: 10233859 PMCID: PMC1866568 DOI: 10.1016/s0002-9440(10)65373-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Our group had previously shown that transfer of the mouse interferon (IFN)-alpha1 gene into the metastasizing TS/A mammary adenocarcinoma resulted in T-cell-mediated tumor rejection and development of antitumor immunity. Moreover, we had shown that the metastatic ability of TS/A tumor cells producing IFN-alpha was strongly impaired, whereas IFN-gamma expression did not influence or augmented metastasis formation by TS/A cells. In this study, we have analyzed the in vitro and in vivo behavior of various TS/A tumor cell clones isolated after the transduction with a recombinant retroviral vector carrying the mouse IFN-beta gene. We have also compared the tumorigenicity of these clones with that of TS/A cells expressing IFN-alpha1. BALB/c mice were inoculated subcutaneously with parental TS/A cells, transduction control TS/A cells, or TS/A cells producing IFN-alpha or IFN-beta. Tumor growth was evaluated by the measurement of tumor masses and analysis of survival. The features of tumor growth and rejection were examined by histological and immunohistochemical analyses. The metastatic ability of parental TS/A cells, transduction control TS/A cells, or TS/A cells producing IFN-alpha, IFN-beta, or IFN-gamma was evaluated after intravenous injection of the tumor cells into BALB/c mice by counting of the lung metastatic nodules and analysis of survival. A strong inhibition of tumorigenicity and development of tumor immunity were observed upon subcutaneous injection of syngeneic mice with TS/A tumor cells producing high amounts of IFN-beta, but not with clones expressing low levels of the cytokine, as observed for cells expressing IFN-alpha. IFN-alpha secretion by TS/A cells at the site of tumor growth induced a stronger inflammatory response as compared with IFN-beta, which appeared to be more active in the inhibition of tumor-induced angiogenesis. Notably, the metastatic ability of IFN-beta-producing TS/A cells after intravenous injection was either not affected or only slightly impaired as compared with parental TS/A tumor cells. In contrast, even cells producing low levels of IFN-alpha proved to be poorly metastatic. These findings represent the first comparison of the effectiveness of IFN-alpha versus IFN-beta produced by genetically modified cells on their tumorigenic behavior and suggest the existence of some notable differences in the capabilities of these two cytokines to induce a host antitumor reactivity in mice.
Collapse
Affiliation(s)
- C Rozera
- Laboratory of Virology, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
36
|
van Seventer GA, Semnani RT, Palmer EM, McRae BL, van Seventer JM. Integrins and T helper cell activation. Transplant Proc 1998; 30:4270-4. [PMID: 9865361 DOI: 10.1016/s0041-1345(98)01410-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- G A van Seventer
- Department of Pathology, University of Chicago, Illinois 60637-1463, USA
| | | | | | | | | |
Collapse
|
37
|
DaSilva L, Kirken RA, Taub DD, Evans GA, Duhé RJ, Bailey MA, Farrar WL. Molecular cloning of FKHRL1P2, a member of the developmentally regulated fork head domain transcription factor family. Gene 1998; 221:135-42. [PMID: 9852958 DOI: 10.1016/s0378-1119(98)00441-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Here we report the expression of a fork head domain protein in human T helper cells. We cloned and characterized a fork head cDNA from human T helper cell mRNA using differential display RT-PCR. The cDNA contains a 546-nucleotide (nt) open reading frame (ORF) that codes for the carboxyl-terminal 180 amino acids (aa) of the recently identified fkhrl1 gene. This ORF does not contain the characteristic DNA-binding domain found in members of the forkhead protein family. In-vitro transcription/translation of this cDNA expressed a protein of approximately 20 kDa. We have generated antibodies that specifically immunoprecipitated the in-vitro-translated 20-kDa protein. This antibody also recognizes in human T lymphocytes a 70-kDa protein corresponding in size to that predicted for the fkhrl1 gene product. The mRNA levels for fkhrl1 is elevated in T helper-induced lymphocytes in comparison to PHA-stimulated T lymphocytes. Further characterization of FKHRL1 and its related family members should shed light on the transcriptional mechanisms of this fork head gene subfamily and their role in T helper cell differentiation and regulation of cell growth.
Collapse
MESH Headings
- Amino Acid Sequence
- Base Sequence
- Blotting, Northern
- Cell Differentiation
- Cloning, Molecular
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- DNA-Binding Proteins/analysis
- DNA-Binding Proteins/genetics
- Forkhead Box Protein O1
- Forkhead Box Protein O3
- Forkhead Transcription Factors
- Gene Expression Regulation, Developmental
- Humans
- Molecular Sequence Data
- Precipitin Tests
- Protein Biosynthesis
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Sequence Alignment
- Sequence Analysis, DNA
- T-Lymphocytes, Helper-Inducer/chemistry
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Helper-Inducer/metabolism
- Transcription Factors/analysis
- Transcription Factors/genetics
Collapse
Affiliation(s)
- L DaSilva
- Division of Basic Science, IRSP, SAIC Frederick, MD 21702, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Tang L, Benjaponpitak S, DeKruyff RH, Umetsu DT. Reduced prevalence of allergic disease in patients with multiple sclerosis is associated with enhanced IL-12 production. J Allergy Clin Immunol 1998; 102:428-35. [PMID: 9768584 DOI: 10.1016/s0091-6749(98)70131-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND We previously showed that the prevalence of allergic disease is decreased in patients with multiple sclerosis (MS); however, the mechanisms that explain this finding have not previously been defined. OBJECTIVES We have demonstrated that protection of patients with MS from allergic disease may be caused by the production in monocytes from these patients of elevated quantities of IL-12 compared with that observed in monocytes from individuals with allergies. METHODS Purified monocytes from peripheral blood of subjects with or without allergies and from individuals with MS were directly stimulated with Staphylococcus aureus Cowan strain I in the absence of T cells. IL-12 was quantitated by a sensitive reverse transcription, competitive PCR. RESULTS IL-12 production was 5-fold greater in monocytes from patients with MS (n = 11) than that from individuals with allergies (n = 10) (for subjects with MS, 1.90+/-0.18 vs 1.24+/-0.19 log10 fmol/microL for individuals with allergies) (P = .02). Although the production of IL-12 in monocytes from patients with MS was slightly higher than that from subjects without allergies, this difference was not statistically significant. CONCLUSIONS IL-12 production in individuals with MS is much greater than in individuals with allergies. Because IL-12 induces TH1 cytokine synthesis and reduces the production of TH2 cytokines, which amplify and prolong allergic inflammation, these studies suggest that enhanced IL-12 production may protect individuals with MS from the development of allergy but may predispose such individuals toward autoimmune inflammation in the central nervous system.
Collapse
Affiliation(s)
- L Tang
- Department of Pediatrics, Stanford University, Calif 94305-5208, USA
| | | | | | | |
Collapse
|
39
|
Abstract
Multiple sclerosis is an inflammatory demyelinating disease of the central nervous system that is of possible autoimmune origin. This article is divided into three parts, reviewing recent advances in three selected topics regarding the immunology of multiple sclerosis. The first part addresses the consequences of T cell and oligodendrocyte death in the inflammatory lesions. The second covers the recent experimental evidence favouring the involvement of infectious agents in the pathophysiology of central nervous system autoimmune diseases. The third part concerns the mode of action of interferon-beta in multiple sclerosis. These new advances have lead to a better understanding of the immunopathology of multiple sclerosis and therefore open new therapeutic possibilities.
Collapse
Affiliation(s)
- R S Liblau
- Department of Neurology, INSERM CJF9608, Salpetriere Hospital, Paris, France
| | | |
Collapse
|
40
|
Frey M, Packianathan NB, Fehniger TA, Ross ME, Wang WC, Stewart CC, Caligiuri MA, Evans SS. Differential Expression and Function of L-Selectin on CD56bright and CD56dim Natural Killer Cell Subsets. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.1.400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
NK cells are the first line of defense against foreign cells, virally infected cells, and tumors. The mechanisms whereby NK cells accumulate in extralymphoid sites in response to pathogenic stimuli are not well understood. The L-selectin adhesion molecule (CD62L) plays a primary role in mediating the initial interaction of leukocytes with vascular endothelium, a crucial step in the extravasation of immune effector cells into tissues. In this report, we show L-selectin to be uniquely expressed on a subset of resting human NK cells (CD56bright). Notably, CD56bright NK cells expressed L-selectin at a higher density than all other peripheral blood leukocytes. NK activation by PMA, IL-2, IL-15, or TGF-β down-regulated L-selectin on the CD56bright subset, while increased L-selectin levels were observed in both the CD56bright and CD56dim NK subsets in response to IL-12, IL-10, or IFN-α. Moreover, CD56bright NK cells bound with high efficiency to physiologic L-selectin ligands on peripheral lymph node high endothelial venules (HEV). In sharp contrast, CD56dim NK cells adhered poorly to HEV and were predominantly L-selectin− or expressed L-selectin only at low density. In CD56bright cells and a subpopulation of CD56dim cells, L-selectin ligation by mAb cross-linking activated lymphocyte function-associated Ag 1 (LFA-1), a second adhesion molecule required for leukocyte extravasation. LFA-1 was expressed on both NK subsets, although its density was constitutively higher on CD56dim cells. Taken together, evidence of differential expression of L-selectin and LFA-1 on CD56bright and CD56dim NK subsets strongly suggests unique migratory properties and functions of these cells during the early immune response to foreign pathogens.
Collapse
Affiliation(s)
| | | | - Todd A. Fehniger
- *Molecular Immunology,
- †Medicine, and
- §Department of Hematology/Oncology, Arthur G. James Cancer Hospital and Research Institute, Ohio State University, Columbus, OH 43210
| | | | | | | | - Michael A. Caligiuri
- *Molecular Immunology,
- †Medicine, and
- §Department of Hematology/Oncology, Arthur G. James Cancer Hospital and Research Institute, Ohio State University, Columbus, OH 43210
| | | |
Collapse
|
41
|
van Wely CA, Blanchard AD, Britten CJ. Differential expression of alpha3 fucosyltransferases in Th1 and Th2 cells correlates with their ability to bind P-selectin. Biochem Biophys Res Commun 1998; 247:307-11. [PMID: 9642121 DOI: 10.1006/bbrc.1998.8786] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the key control points in the trafficking of the T cell effector subsets, Th1 and Th2, to sites of inflammation is their migration out of the bloodstream. The mechanism by which the cells initially adhere to the endothelium is dependent on the selectin family of adhesion molecules. Only polarised Th1 cells are capable of binding P-selectin despite both Th1 and Th2 cells expressing PSGL-1, the P-selectin ligand. This may be due to a secondary modification of PSGL-1 that is present on Th1 but not Th2 cells. One key modification of PSGL-1 is the alpha3 fucosylation of the O-glycans. To address whether the binding of Th1 and Th2 cells may be regulated by fucosylation, we have studied the expression of the alpha3 fucosyltransferases, FucT-IV and VII, using in vitro differentiated mouse T cells. Messenger RNA levels for both FucT-IV and VII were found to be higher in Th1 than Th2 cells. alpha3 fucosyltransferase enzyme activities were also elevated in Th1 cells. The increased expression of the alpha3 fucosyltransferases in Th1 cells correlated with the ability of Th1, but not Th2, cells to bind to P-selectin. Thus, the regulation of the binding of effector T cells to the endothelium, and subsequent trafficking to inflammatory sites, may be controlled by the fucosylation state of PSGL-1 mediated by the selective expression of the alpha3 fucosyltransferases.
Collapse
Affiliation(s)
- C A van Wely
- Immunology Research Unit, Glycobiology Research Unit, Glaxo Wellcome Research and Development Ltd., Gunnels Wood Road, Stevenage, Hertfordshire, United Kingdom
| | | | | |
Collapse
|
42
|
McRae BL, Semnani RT, Hayes MP, van Seventer GA. Type I IFNs Inhibit Human Dendritic Cell IL-12 Production and Th1 Cell Development. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.9.4298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
We have investigated the role of type I IFNs (IFN-α and -β) in human T cell differentiation using anti-CD3 mAb and allogeneic, in vitro-derived dendritic cells (DC) as APCs. DC were very efficient activators of naive CD4+ T cells, providing necessary costimulation and soluble factors to support Th1 differentiation and expansion. Addition of IFN-αβ to DC/T cell cultures resulted in induction of T cell IL-10 production and inhibition of IFN-γ, TNF-α, and LT secretion. Diminished T cell IFN-γ production correlated with IFN-αβ-mediated inhibition of the p40 chain of the IL-12 heterodimer secreted by DC. Suppression of p40 IL-12 and IFN-γ was not due to increased levels of IL-10 in these cultures, and production of IFN-γ could be restored by exogenous IL-12. These data indicate that type I IFNs inhibit DC p40 IL-12 expression, which is required for development of IFN-γ-producing CD4+ T cells. Furthermore, when T cells were restimulated without IFN-β, these cells induced less p40 IL-12 from DC, suggesting that the functional properties of T cells may regulate DC function. Thus, IFN-αβ inhibits both IL-12-dependent and independent Th1 cytokine production and provides a mechanism for inhibition of IL-12-mediated immunity in viral infections.
Collapse
Affiliation(s)
- Bradford L. McRae
- *Committee on Immunology, Department of Pathology, Division of Biological Sciences, University of Chicago, Chicago, IL; and
| | - Roshanak Tolouei Semnani
- *Committee on Immunology, Department of Pathology, Division of Biological Sciences, University of Chicago, Chicago, IL; and
| | - Mark P. Hayes
- †Division of Cytokine Biology, Food and Drug Administration, Bethesda, MD
| | - Gijs A. van Seventer
- *Committee on Immunology, Department of Pathology, Division of Biological Sciences, University of Chicago, Chicago, IL; and
| |
Collapse
|