1
|
Manzanares-Meza LD, Gutiérrez-Román CI, Jiménez-Pineda A, Castro-Martínez F, Patiño-López G, Rodríguez-Arellano E, Valle-Rios R, Ortíz-Navarrete VF, Medina-Contreras O. IL-36γ is secreted through an unconventional pathway using the Gasdermin D and P2X7R membrane pores. Front Immunol 2022; 13:979749. [PMID: 36059446 PMCID: PMC9434278 DOI: 10.3389/fimmu.2022.979749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/04/2022] [Indexed: 11/20/2022] Open
Abstract
Mucosal innate immunity functions as the first line of defense against invading pathogens. Members of the IL-1 family are key cytokines upregulated in the inflamed mucosa. Inflammatory cytokines are regulated by limiting their function and availability through their activation and secretion mechanisms. IL-1 cytokines secretion is affected by the lack of a signal peptide on their sequence, which prevents them from accessing the conventional protein secretion pathway; thus, they use unconventional protein secretion pathways. Here we show in mouse macrophages that LPS/ATP stimulation induces cytokine relocalization to the plasma membrane, and conventional secretion blockade using monensin or Brefeldin A triggers no IL-36γ accumulation within the cell. In silico modeling indicates IL-36γ can pass through both the P2X7R and Gasdermin D pores, and both IL-36γ, P2X7R and Gasdermin D mRNA are upregulated in inflammation; further, experimental blockade of these receptors’ limits IL-36γ release. Our results demonstrate that IL-36γ is secreted mainly by an unconventional pathway through membrane pores formed by P2X7R and Gasdermin D.
Collapse
Affiliation(s)
- Laura D. Manzanares-Meza
- Mexico Children’s Hospital, Endocrinology, Epidemiology & Nutrition Research Unit, Mexico City, Mexico
- Department of Molecular Biomedicine, Center for Research and Advanced Studies, National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | | | - Albertana Jiménez-Pineda
- Mexico Children’s Hospital, Endocrinology, Epidemiology & Nutrition Research Unit, Mexico City, Mexico
| | - Felipe Castro-Martínez
- Mexico Children’s Hospital, Endocrinology, Epidemiology & Nutrition Research Unit, Mexico City, Mexico
| | - Genaro Patiño-López
- Hospital Infantil de Mexico, Unidad de Investigación en Inmunología y Proteómica, Mexico City, Mexico
| | | | - Ricardo Valle-Rios
- Hospital Infantil de Mexico, Unidad de Investigación en Inmunología y Proteómica, Mexico City, Mexico
- Unidad Universitaria de Investigación, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Vianney F. Ortíz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies, National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | - Oscar Medina-Contreras
- Mexico Children’s Hospital, Endocrinology, Epidemiology & Nutrition Research Unit, Mexico City, Mexico
- *Correspondence: Oscar Medina-Contreras,
| |
Collapse
|
2
|
Yang HY, Lin FZ, Yang HW, Yu PL, Huang SM, Chen YC, Tsai CS, Lin CY. The effect of Sirt1 deficiency on Ca 2+ and Na + regulation in mouse ventricular myocytes. J Cell Mol Med 2020; 24:6762-6772. [PMID: 32342656 PMCID: PMC7299725 DOI: 10.1111/jcmm.15327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/06/2020] [Accepted: 04/12/2020] [Indexed: 12/19/2022] Open
Abstract
This study addressed the hypothesis that cardiac Sirtuin 1 (Sirt1) deficiency alters cardiomyocyte Ca2+ and Na+ regulation, leading to cardiac dysfunction and arrhythmogenesis. We used mice with cardiac‐specific Sirt1 knockout (Sirt1−/−). Sirt1flox/flox mice were served as control. Sirt1−/− mice showed impaired cardiac ejection fraction with increased ventricular spontaneous activity and burst firing compared with those in control mice. The arrhythmic events were suppressed by KN93 and ranolazine. Reduction in Ca2+ transient amplitudes and sarcoplasmic reticulum (SR) Ca2+ stores, and increased SR Ca2+ leak were shown in the Sirt1−/− mice. Electrophysiological measurements were performed using patch‐clamp method. While L‐type Ca2+ current (ICa, L) was smaller in Sirt1−/− myocytes, reverse‐mode Na+/Ca2+ exchanger (NCX) current was larger compared with those in control myocytes. Late Na+ current (INa, L) was enhanced in the Sirt1−/− mice, alongside with elevated cytosolic Na+ level. Increased cytosolic and mitochondrial reactive oxygen species (ROS) were shown in Sirt1−/− mice. Sirt1−/− cardiomyocytes showed down‐regulation of L‐type Ca2+ channel α1c subunit (Cav1.2) and sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a), but up‐regulation of Ca2+/calmodulin‐dependent protein kinase II and NCX. In conclusions, these findings suggest that deficiency of Sirt1 impairs the regulation of intracellular Ca2+ and Na+ in cardiomyocytes, thereby provoking cardiac dysfunction and arrhythmogenesis.
Collapse
Affiliation(s)
- Hsiang-Yu Yang
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Feng-Zhi Lin
- Grade institute of life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hui-Wen Yang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Pei-Ling Yu
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Yuan Lin
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
3
|
Sitia R, Rubartelli A. Evolution, role in inflammation, and redox control of leaderless secretory proteins. J Biol Chem 2020; 295:7799-7811. [PMID: 32332096 DOI: 10.1074/jbc.rev119.008907] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Members of the interleukin (IL)-1 family are key determinants of inflammation. Despite their role as intercellular mediators, most lack the leader peptide typically required for protein secretion. This lack is a characteristic of dozens of other proteins that are actively and selectively secreted from living cells independently of the classical endoplasmic reticulum-Golgi exocytic route. These proteins, termed leaderless secretory proteins (LLSPs), comprise proteins directly or indirectly involved in inflammation, including cytokines such as IL-1β and IL-18, growth factors such as fibroblast growth factor 2 (FGF2), redox enzymes such as thioredoxin, and proteins most expressed in the brain, some of which participate in the pathogenesis of neurodegenerative disorders. Despite much effort, motifs that promote LLSP secretion remain to be identified. In this review, we summarize the mechanisms and pathophysiological significance of the unconventional secretory pathways that cells use to release LLSPs. We place special emphasis on redox regulation and inflammation, with a focus on IL-1β, which is secreted after processing of its biologically inactive precursor pro-IL-1β in the cytosol. Although LLSP externalization remains poorly understood, some possible mechanisms have emerged. For example, a common feature of LLSP pathways is that they become more active in response to stress and that they involve several distinct excretion mechanisms, including direct plasma membrane translocation, lysosome exocytosis, exosome formation, membrane vesiculation, autophagy, and pyroptosis. Further investigations of unconventional secretory pathways for LLSP secretion may shed light on their evolution and could help advance therapeutic avenues for managing pathological conditions, such as diseases arising from inflammation.
Collapse
Affiliation(s)
- Roberto Sitia
- Division of Genetics and Cell Biology, Protein Transport and Secretion Unit, IRCCS Ospedale San Raffaele/Università Vita-Salute San Raffaele, Milan, Italy
| | - Anna Rubartelli
- Division of Genetics and Cell Biology, Protein Transport and Secretion Unit, IRCCS Ospedale San Raffaele/Università Vita-Salute San Raffaele, Milan, Italy .,Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
4
|
Ge W, Li D, Gao Y, Cao X. The Roles of Lysosomes in Inflammation and Autoimmune Diseases. Int Rev Immunol 2014; 34:415-31. [DOI: 10.3109/08830185.2014.936587] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
5
|
Dinarello CA, Kaplanski G. Interleukin-18 treatment options for inflammatory diseases. Expert Rev Clin Immunol 2014; 1:619-32. [DOI: 10.1586/1744666x.1.4.619] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
6
|
Abstract
Unlike most cytokines, IL-1β lacks a secretory signal sequence raising the question of how is this cytokine processed and delivered outside the producing cells. After the seminal observation that IL-1β is actively secreted by human monocytes through a route alternative to the classic endoplasmic reticulum-Golgi, several different pathways have been proposed for IL-1β secretion in different cell types and culture conditions, some of which are unique to macrophage cell lines. Here we describe the most credited of these pathways. In particular, we will focus on IL-1β secretion from primary human blood monocytes. In fact, although data from macrophages or macrophage cell lines are predominant, secretion of IL-1β by monocytes is the most clinically relevant.
Collapse
|
7
|
Monobe M, Ema K, Tokuda Y, Maeda-Yamamoto M. Green tea catechin induced phagocytosis can be blocked by catalase and an inhibitor of transient receptor potential melastatin 2 (TRPM2). Cytotechnology 2013; 66:561-6. [PMID: 23896702 DOI: 10.1007/s10616-013-9618-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 07/15/2013] [Indexed: 12/31/2022] Open
Abstract
The major polyphenols in green tea, (-)-epigallocatechin and (-)-epigallocatechin gallate, have been shown to enhance the phagocytic activity of macrophage-like cells; however, the mechanism involved was not clarified. In this study, we have identified that the catechins induced phagocytosis can be blocked by catalase and an inhibitor of transient receptor potential melastatin 2.
Collapse
Affiliation(s)
- Manami Monobe
- NARO Institute of Vegetable and Tes Science, 2769 Kanaya-Shishidoi, Shimada, Shizuoka, 428-8501, Japan,
| | | | | | | |
Collapse
|
8
|
Carta S, Lavieri R, Rubartelli A. Different Members of the IL-1 Family Come Out in Different Ways: DAMPs vs. Cytokines? Front Immunol 2013; 4:123. [PMID: 23745123 PMCID: PMC3662868 DOI: 10.3389/fimmu.2013.00123] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 05/08/2013] [Indexed: 11/13/2022] Open
Abstract
Intercellular communications control fundamental biological processes required for the survival of multicellular organisms. Secretory proteins are among the most important messengers in this network of information. Proteins destined to the extracellular environment contain a signal sequence with the necessary information to target them to the Endoplasmic Reticulum, and are released by a "classical" pathway of secretion. However, in the early 1990s it became evident that non-classical mechanisms must exist for the secretion of some proteins, which in spite of their extracellular localization and function, lack a signal peptide. Indeed, the group of leaderless secretory proteins rapidly grew and is still growing. Many of them are implicated in the regulation of the inflammatory response. Interestingly, most members of the IL-1 family (IL-1F), including the master pro-inflammatory cytokine IL-1β, are leaderless proteins and find their way out of the cells in different manners. In this article, we will review current hypotheses on the mechanisms of externalization of IL-1F members and discuss their relevance with respect to the different functions (as cytokines or as DAMPs) played by the different IL-1 proteins.
Collapse
Affiliation(s)
- Sonia Carta
- Cell Biology Unit, IRCSS Azienda Ospedale Università San Martino-IST , Genoa , Italy
| | | | | |
Collapse
|
9
|
Gulinelli S, Salaro E, Vuerich M, Bozzato D, Pizzirani C, Bolognesi G, Idzko M, Di Virgilio F, Ferrari D. IL-18 associates to microvesicles shed from human macrophages by a LPS/TLR-4 independent mechanism in response to P2X receptor stimulation. Eur J Immunol 2012; 42:3334-45. [PMID: 22996386 DOI: 10.1002/eji.201142268] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 07/10/2012] [Accepted: 09/12/2012] [Indexed: 11/06/2022]
Abstract
Extracellular ATP, released upon microbial infection, cell damage, or inflammation, acts as an alert signal toward immune cells by activating P2 receptors. The nucleotide causes microvesicle (MV) shedding from immune and nonimmune cells. Here, we show that IL-18 associates with MVs shed by human ex vivo macrophages upon P2X receptor stimulation. MV shedding was potently induced by ATP and by the P2X7 agonist 3'-benzoylbenzoyl adenosine 5'-triphosphate, while it was greatly reduced by P2X irreversible inhibitor-oxidized ATP and by the specific P2X7 inhibitors KN-62, A-740003, and A-438079. Peculiarly, the P2X7 subtype was highly present in the MVs, while on the contrary the P2X3 and P2X4 subtypes were almost absent. The Ca(2+) ionophore A23187 mimicked the effect of 3'-benzoylbenzoyl adenosine 5'-triphosphate suggesting that an intracellular Ca(2+) increase was sufficient to evoke MV shedding. Caspase inhibitors Ac-YVAD-CMK or Z-YVAD-CMK did not block the cleavage of MV-associated pro-IL-18. Pro-IL-18 formation in macrophages did not require pretreatment of cells with LPS, as the procytokine was already present in unprimed macrophages and did not decrease by incubating cells with the LPS-binding antibiotic polymyxin B nor with the TLR-4 intracellular inhibitor CLI-095. These data reveal a nucleotide-based mechanism responsible for the shedding of MV to which IL-18 is associated.
Collapse
Affiliation(s)
- Sara Gulinelli
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, University of Ferrara, Ferrara, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Sutinen EM, Pirttilä T, Anderson G, Salminen A, Ojala JO. Pro-inflammatory interleukin-18 increases Alzheimer's disease-associated amyloid-β production in human neuron-like cells. J Neuroinflammation 2012; 9:199. [PMID: 22898493 PMCID: PMC3458954 DOI: 10.1186/1742-2094-9-199] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 07/28/2012] [Indexed: 11/10/2022] Open
Abstract
Background Alzheimer’s disease (AD) involves increased accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles as well as neuronal loss in various regions of the neocortex. Neuroinflammation is also present, but its role in AD is not fully understood. We previously showed increased levels of pro-inflammatory cytokine interleukin-18 (IL-18) in different regions of AD brains, where it co-localized with Aβ-plaques, as well as the ability of IL-18 to increase expression of glycogen synthase kinase-3β (GSK-3β) and cyclin dependent kinase 5, involved in hyperphosphorylation of tau-protein. Elevated IL-18 has been detected in several risk conditions for AD, including obesity, type-II diabetes, and cardiovascular diseases as well as in stress. Methods We differentiated SH-SY5Y neuroblastoma cells as neuron-like and exposed them to IL-18 for various times. We examined the protein levels of amyloid-β precursor protein (APP) and its processing products, its cleaving enzymes, involved in amyloidogenic processing of APP, and markers of apoptosis. Results IL-18 increased protein levels of the β-site APP-cleaving enzyme BACE-1, the N-terminal fragment of presenilin-1 and slightly presenilin enhancer 2, both of which are members of the γ-secretase complex, as well as Fe65, which is a binding protein of the C-terminus of APP and one regulator for GSK-3β. IL-18 also increased APP expression and phosphorylation, which preceded increased BACE-1 levels. Further, IL-18 altered APP processing, increasing Aβ40 production in particular, which was inhibited by IL-18 binding protein. Increased levels of soluble APPβ were detected in culture medium after the IL-18 exposure. IL-18 also increased anti-apoptotic bcl-xL levels, which likely counteracted the minor increase of the pro-apoptotic caspase-3. Lactate dehydrogenase activity in culture medium was unaffected. Conclusions The IL-18 induction of BACE-1, APP processing, and Aβ is likely to be linked to stress-associated adaptations in neurons during the course of normal functioning and development. However, in the course of wider changes in the aging brain, and particularly in AD, the effects of heightened or prolonged levels of IL-18 may contribute to the process of AD, including via increased Aβ.
Collapse
Affiliation(s)
- Elina M Sutinen
- University of Eastern Finland, Institute of Clinical Medicine/ Neurology, Canthia, FI-70211, Kuopio, Finland
| | | | | | | | | |
Collapse
|
11
|
Shumilina E, Huber SM, Lang F. Ca2+ signaling in the regulation of dendritic cell functions. Am J Physiol Cell Physiol 2011; 300:C1205-14. [PMID: 21451105 DOI: 10.1152/ajpcell.00039.2011] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Dendritic cells (DCs) are highly versatile antigen-presenting cells critically involved in both innate and adaptive immunity as well as maintenance of self-tolerance. DC function is governed by Ca(2+) signaling, which directs the DC responses to diverse antigens, including Toll-like receptor ligands, intact bacteria, and microbial toxins. Ca(2+)-sensitive DC functions include DC activation, maturation, migration, and formation of immunological synapses with T cells. Moreover, alterations of cytosolic Ca(2+) trigger immune suppression or switch off DC activity. Ca(2+) signals are generated by the orchestration of Ca(2+) transport processes across plasma, endoplasmic reticulum, and inner mitochondrial membrane. These processes include active pumping of Ca(2+), Ca(2+)/Na(+) antiport, and electrodiffusion through Ca(2+)-permeable channels or uniporters. Ca(2+) channels in the plasma membrane such as Ca(2+) release-activated Ca(2+) or L-type Ca(2+) channels are tightly regulated by the membrane potential which in turn depends on the activity of voltage-gated K(+) or Ca(2+)-activated nonselective cation channels. The rapidly growing knowledge on the function and regulation of these membrane transport proteins provides novel insight into pathophysiological mechanisms underlying dysfunction of the immune system and opens novel therapeutic opportunity to favorably influence the function of the immune system.
Collapse
Affiliation(s)
- Ekaterina Shumilina
- Department of Physiology, University of Tübingen, Gmelinstrasse 5, Tübingen, Germany.
| | | | | |
Collapse
|
12
|
Antigen-specific T-cell responses to a recombinant fowlpox virus are dependent on MyD88 and interleukin-18 and independent of Toll-like receptor 7 (TLR7)- and TLR9-mediated innate immune recognition. J Virol 2011; 85:3385-96. [PMID: 21248035 DOI: 10.1128/jvi.02000-10] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Fowlpox virus (FWPV) is a double-stranded DNA virus long used as a live-attenuated vaccine against poultry diseases, but more recent interest has focused on its use as a mammalian vaccine vector. Here, in a mouse model system using FWPV encoding the nominal target antigen chicken ovalbumin (OVA) (FWPV(OVA)), we describe for the first time some of the fundamental processes by which FWPV engages both the innate and adaptive immune systems. We show that Toll-like receptor 7 (TLR7) and TLR9 are important for type I interferon secretion by dendritic cells, while TLR9 is solely required for proinflammatory cytokine secretion. Despite this functional role for TLR7 and TLR9 in vitro, only the adapter protein myeloid differentiation primary response gene 88 (MyD88) was shown to be essential for the formation of adaptive immunity to FWPV(OVA) in vivo. The dependence on MyD88 was confined only to the T-cell compartment and was not related to its contribution to TLR signaling, dendritic cell maturation, or the capture and presentation of FWPV-derived OVA antigen. We demonstrate that this is not by means of mediating T-cell-dependent interleukin-1 (IL-1) signaling, but rather, we suggest that MyD88 functions to support T-cell-specific IL-18 receptor signaling, which in turn is essential for the formation of adaptive immunity to FWPV-encoded OVA.
Collapse
|
13
|
Orengo AM, Fabbi M, Miglietta L, Andreani C, Bruzzone M, Puppo A, Cristoforoni P, Centurioni MG, Gualco M, Salvi S, Boccardo S, Truini M, Piazza T, Canevari S, Mezzanzanica D, Ferrini S. Interleukin (IL)-18, a biomarker of human ovarian carcinoma, is predominantly released as biologically inactive precursor. Int J Cancer 2011; 129:1116-25. [PMID: 21710494 DOI: 10.1002/ijc.25757] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 10/13/2010] [Indexed: 12/12/2022]
Abstract
Interleukin (IL)-18 is a proinflammatory and immune-enhancing cytokine, which exerts antitumor effects in vivo, mediated by the induction of interferon (IFN)γ. We previously reported that IL-18 processing is defective in epithelial ovarian carcinoma (EOC) cells, which secrete an inactive precursor (pro-IL-18) in vitro. In addition, IL-18 was reported as a potential biomarker of EOC. Here, we further investigated its role as a serological marker in human EOC and addressed its possible biological activity in vivo. Our data indicate that immunoreactive IL-18 is increased in EOC patients' sera at diagnosis as compared with age-matched healthy women. IL-18 levels were higher in the ascitic fluids than in sera, suggesting a local production in the peritoneal cavity. Indeed, immunohistochemical analysis of tumors showed IL-18 expression in cytokeratine-positive neoplastic cells, although also scattered histiocytes and some lymphoid cells stained for IL-18. The detection of human IL-18 in sera and ascitic fluids of immunodeficient mice, orthotopically implanted with human EOC cells, further suggested that circulating IL-18 is tumor-derived. However, IL-18 is not an EOC specific biomarker, as increased serum levels were found also in some endometrial cancer patients. By means of a new monoclonal antibody, we characterized IL-18 present in the ascitic fluid as pro-IL-18, which is biologically inactive. Accordingly, IFNγ was not increased in EOC patients' sera and ascitic fluids and showed no correlation with IL-18 levels. Altogether these data indicate that IL-18 in EOC fluids is predominantly tumor-derived and that its lack of biological activity may represent a mechanism of tumor-escape.
Collapse
Affiliation(s)
- Anna Maria Orengo
- Department of Translational Oncology, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Rehm KE, Connor RF, Jones GJB, Yimbu K, Mannie MD, Roper RL. Vaccinia virus decreases major histocompatibility complex (MHC) class II antigen presentation, T-cell priming, and peptide association with MHC class II. Immunology 2010; 128:381-92. [PMID: 20067538 DOI: 10.1111/j.1365-2567.2009.03120.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Vaccinia virus (VACV) is the current live virus vaccine used to protect humans against smallpox and monkeypox, but its use is contraindicated in several populations because of its virulence. It is therefore important to elucidate the immune evasion mechanisms of VACV. We found that VACV infection of antigen-presenting cells (APCs) significantly decreased major histocompatibility complex (MHC) II antigen presentation and decreased synthesis of 13 chemokines and cytokines, suggesting a potent viral mechanism for immune evasion. In these model systems, responding T cells were not directly affected by virus, indicating that VACV directly affects the APC. VACV significantly decreased nitric oxide production by peritoneal exudate cells and the RAW macrophage cell line in response to lipopolysaccharide (LPS) and interferon (IFN)-gamma, decreased class II MHC expression on APCs, and induced apoptosis in macrophages and dendritic cells. However, VACV decreased antigen presentation by 1153 B cells without apparent apoptosis induction, indicating that VACV differentially affects B lymphocytes and other APCs. We show that the key mechanism of VACV inhibition of antigen presentation may be its reduction of antigenic peptide loaded into the cleft of MHC class II molecules. These data indicate that VACV evades the host immune response by impairing critical functions of the APC.
Collapse
Affiliation(s)
- Kristina E Rehm
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | | | | | | | | | | |
Collapse
|
15
|
Rehm KE, Connor RF, Jones GJB, Yimbu K, Roper RL. Vaccinia virus A35R inhibits MHC class II antigen presentation. Virology 2009; 397:176-86. [PMID: 19954808 DOI: 10.1016/j.virol.2009.11.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 10/05/2009] [Accepted: 11/05/2009] [Indexed: 10/20/2022]
Abstract
The Vaccinia virus gene A35R (Copenhagen designation) is highly conserved in mammalian-tropic poxviruses and is an important virulence factor, but its function was unknown. We show herein that A35 does not affect viral infectivity, apoptosis induction, or replication; however, we found that A35 significantly inhibited MHC class II-restricted antigen presentation, immune priming of T lymphocytes, and subsequent chemokine and cytokine synthesis. A35 localized to endosomes and reduced the amount of a model antigenic peptide displayed in the cleft of class II MHC. In addition, A35 decreased VV specific T cell responses in vivo. Thus, this is the first report identifying a function for the A35 protein in virulence as well as the first report identifying a VV gene that inhibits peptide antigen presentation.
Collapse
Affiliation(s)
- Kristina E Rehm
- Brody School of Medicine at East Carolina University, Department of Microbiology and Immunology, Greenville, NC, USA
| | | | | | | | | |
Collapse
|
16
|
Anthrax lethal toxin triggers the formation of a membrane-associated inflammasome complex in murine macrophages. Infect Immun 2009; 77:1262-71. [PMID: 19124602 DOI: 10.1128/iai.01032-08] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Multiple microbial components trigger the formation of an inflammasome complex that contains pathogen-specific nucleotide oligomerization and binding domain (NOD)-like receptors (NLRs), caspase-1, and in some cases the scaffolding protein ASC. The NLR protein Nalp1b has been linked to anthrax lethal toxin (LT)-mediated cytolysis of murine macrophages. Here we demonstrate that in unstimulated J774A.1 macrophages, caspase-1 and Nalp1b are membrane associated and part of approximately 200- and approximately 800-kDa complexes, respectively. LT treatment of these cells resulted in caspase-1 recruitment to the Nalp1b-containing complex, concurrent with processing of cytosolic caspase-1 substrates. We further demonstrated that Nalp1b and caspase-1 are able to interact with each other. Intriguingly, both caspase-1 and Nalp1b were membrane associated, while the caspase-1 substrate interleukin-18 was cytosolic. Caspase-1-associated inflammasome components included, besides Nalp1b, proinflammatory caspase-11 and the caspase-1 substrate alpha-enolase. Asc was not part of the Nalp1b inflammasome in LT-treated macrophages. Taken together, our findings suggest that LT triggers the formation of a membrane-associated inflammasome complex in murine macrophages, resulting in cleavage of cytosolic caspase-1 substrates and cell death.
Collapse
|
17
|
Devogelaere B, Sammels E, De Smedt H. The IRBIT domain adds new functions to the AHCY family. Bioessays 2008; 30:642-52. [PMID: 18536033 DOI: 10.1002/bies.20772] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During the past few years, the IRBIT domain has emerged as an important add-on of S-adenosyl-L-homocystein hydrolase (AHCY), thereby creating the new family of AHCY-like proteins. In this review, we discuss the currently available data on this new family of proteins. We describe the IRBIT domain as a unique part of these proteins and give an overview of its regulation via (de)phosphorylation and proteolysis. The second part of this review is focused on the potential functions of the AHCY-like proteins. We propose that the IRBIT domain serves as an anchor for targeting AHCY-like proteins towards cytoplasmic targets. This leads to regulation of (i) intracellular Ca2+ via the inositol 1,4,5-trisphosphate receptor (IP3R), (ii) intracellular pH via the Na+/HCO3 - cotransporters (NBCs); whereas inactivation of the IRBIT domain induces (iii) nuclear translocation and regulation of AHCY activity. Dysfunction of AHCY-like proteins will disturb these three important functions, with various biological implications.
Collapse
Affiliation(s)
- Benoit Devogelaere
- Laboratory of Molecular and Cellular Signalling, Department of Molecular Cell Biology, Katholieke Universiteit Leuven, Belgium
| | | | | |
Collapse
|
18
|
Ca(2+)- and glycoconjugates-dependent prey capture in the heliozoon Actinophrys sol. Eur J Protistol 2008; 44:163-7. [PMID: 18178392 DOI: 10.1016/j.ejop.2007.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 11/07/2007] [Accepted: 11/08/2007] [Indexed: 11/21/2022]
Abstract
Exocytosis of extrusomes, secretory granules found in protozoa, is involved in prey capture by the heliozoon Actinophrys sol. Here, we show that extracellular Ca(2+) is necessary for exocytosis and prey capture in A. sol. We found that A. sol could not capture prey cells in a Ca(2+)-free solution. L-type Ca(2+) channel blockers and a calmodulin antagonist also inhibited the capture of prey. These results suggest that Ca(2+) influx via L-type Ca(2+) channels plays a crucial role in exocytosis in A. sol. Concanavalin A (Con A) also inhibited prey capture, and the inhibition was relieved by the addition of its hapten sugar, alpha-mannoside, suggesting that Con A-binding glycoconjugates are implicated in exocytosis of extrusomes and the adhesion of prey cells.
Collapse
|
19
|
Sugama S, Conti B. Interleukin-18 and stress. ACTA ACUST UNITED AC 2007; 58:85-95. [PMID: 18295340 DOI: 10.1016/j.brainresrev.2007.11.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 11/02/2007] [Accepted: 11/16/2007] [Indexed: 01/15/2023]
Abstract
Interleukin-18 (IL-18) is a pro-inflammatory cytokine believed to play a role in a variety of conditions and diseases including infections, autoimmunity, cancer, diabetes and atherosclerosis. IL-18 is also a possible contributor to the sickness syndrome by inducing anorexia and sleep. Originally recognized to be produced by cells of the immune system, IL-18 is also found in endocrine tissues, including the adrenal and the pituitary glands, and in the central nervous system where it is produced by microglial and ependymal cells as well as by neurons of the medial habenular nucleus. IL-18 is produced constitutively and its levels can increase during infection but also during stress in the absence of an exogenous stimulus. IL-18 levels are elevated by activation of the hypothalamic-pituitary-adrenal (HPA) axis in a tissue specific way via differential promoter and splicing usage, and may be down-regulated by the activation of the para-sympathetic system. This suggested the possibility that IL-18 may participate in the regulation of the HPA axis or that it may have a role in mediating the CNS dependent effects on the susceptibility to or the progression of diseases. This review summarizes the evidence linking stress and IL-18 and discusses the possible implication of the neuro-immuno-modulatory action of IL-18.
Collapse
Affiliation(s)
- Shuei Sugama
- Department of Physiology, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan.
| | | |
Collapse
|
20
|
Barr DP, Belz GT, Reading PC, Wojtasiak M, Whitney PG, Heath WR, Carbone FR, Brooks AG. A role for plasmacytoid dendritic cells in the rapid IL-18-dependent activation of NK cells following HSV-1 infection. Eur J Immunol 2007; 37:1334-42. [PMID: 17407097 PMCID: PMC2699043 DOI: 10.1002/eji.200636362] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Natural killer (NK) cells play a crucial role in the initial response to viral infections but the mechanisms controlling their activation are unclear. We show a rapid and transient activation of NK cells that results in the production of IFN-gamma immediately following infection with herpes simplex virus type 1 (HSV-1). Activation of NK cells leading to synthesis of IFN-gamma was not mediated by a direct interaction with virus but required the presence of additional cell types and was largely dependent on the cytokine IL-18, but not IL-12. HSV-1-induced IFN-gamma expression by NK cells in vitro was impaired in spleen cultures depleted of CD11c(+) cells. Conversely, coculture of NK cells with virus-exposed conventional DC or plasmacytoid (p)DC restored the production of IFN-gamma, indicating that multiple DC subsets could mediate NK cell activation. While conventional DC populations stimulated NK cells independently of IL-18, they were less effective than pDC in promoting NK cell IFN-gamma expression. In contrast, the potent stimulation of NK cells by pDC was dependent on IL-18 as pDC from IL-18-deficient mice only activated a similar proportion of NK cells as conventional DC. These data identify IL-18 as a crucial factor for pDC-mediated NK cell regulation.
Collapse
Affiliation(s)
- Daniel P Barr
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Several autoimmune diseases are thought to be mediated in part by interleukin (IL)-18. Many are those with associated increased interferon-gamma (IFNgamma) levels such as systemic lupus erythematosus, macrophage activation syndrome, rheumatoid arthritis, Crohn's disease, psoriasis, and graft-versus-host disease. In addition, ischemia, including acute renal failure in human beings, appears to involve IL-18. Animal studies also support the concept that IL-18 is a key player in models of lupus erythematosus, atherosclerosis, graft-versus-host disease, and hepatitis. Unexpectedly, IL-18 plays a role in appetite control and the development of obesity. IL-18 is a member of the IL-1 family; IL-1beta and IL-18 are related closely, and both require the intracellular cysteine protease caspase-1 for biological activity. The IL-18 binding protein, a naturally occurring and specific inhibitor of IL-18, neutralizes IL-18 activities and has been shown to be safe in patients. Other options for reducing IL-18 activities are inhibitors of caspase-1, human monoclonal antibodies to IL-18, soluble IL-18 receptors, and anti-IL-18 receptor monoclonal antibodies.
Collapse
Affiliation(s)
- Charles A Dinarello
- Department of Medicine, Division of Infectious Diseases, University of Colorado Health Sciences Center, Denver, CO, USA
| |
Collapse
|
22
|
Semino C, Ceccarelli J, Lotti LV, Torrisi MR, Angelini G, Rubartelli A. The maturation potential of NK cell clones toward autologous dendritic cells correlates with HMGB1 secretion. J Leukoc Biol 2006; 81:92-9. [PMID: 16997859 DOI: 10.1189/jlb.0306172] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Interaction of NK cells with autologous immature dendritic cells (iDCs) results in reciprocal activation. We have previously reported that NK cells trigger iDC to polarize and secrete IL-18; in turn, DC-activated NK cells secrete the nuclear protein/proinflammatory cytokine high mobility group box protein 1 (HMGB1), which induces DC maturation and prevents DC from lysis. However, activated NK cells can also kill iDC. To investigate whether effector and maturative properties may coexist or segregate in different NK subsets, human NK cell clones were generated and analyzed for their effects on iDC. We found that the ability of different NK cell clones to induce iDC maturation is unlinked to their phenotypic and cytolytic features but correlates with the relocation of HMGB1 from nucleus to cytoplasm. "Maturative" NK cell clones secrete HMGB1 spontaneously. It is interesting that secretion is strongly enhanced by engagement of the surface molecule NKp30 but only slightly induced by triggering of the activating NK receptor CD16. However, culturing freshly isolated NK cells for 1 week with low doses of anti-CD16 triggers the relocation of HMGB1 from nucleus to cytoplasm and its spontaneous secretion, resulting in a stronger maturation potential of the NK cells. Together, our data indicate that NK cells comprise functionally different subsets, endowed with different capacities to secrete HMGB1 and to induce maturation of autologous iDC. Nonetheless, maturation properties can be modulated by different stimuli. This suggests that depending on the environmental stimuli, NK/iDC interaction can lead to different outcomes, thus influencing immune response.
Collapse
Affiliation(s)
- Claudia Semino
- Laboratory of Experimental Oncology E, Department of Translational Oncology, National Cancer Research Institute, Largo R. Benzi, 10, 16132 Genova, Italy
| | | | | | | | | | | |
Collapse
|
23
|
Goth SR, Chu RA, Gregg JP, Cherednichenko G, Pessah IN. Uncoupling of ATP-mediated calcium signaling and dysregulated interleukin-6 secretion in dendritic cells by nanomolar thimerosal. ENVIRONMENTAL HEALTH PERSPECTIVES 2006; 114:1083-91. [PMID: 16835063 PMCID: PMC1513334 DOI: 10.1289/ehp.8881] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Dendritic cells (DCs) , a rare cell type widely distributed in the soma, are potent antigen-presenting cells that initiate primary immune responses. DCs rely on intracellular redox state and calcium (Ca2+) signals for proper development and function, but the relationship between these two signaling systems is unclear. Thimerosal (THI) is a mercurial used to preserve vaccines and consumer products, and is used experimentally to induce Ca2+ release from microsomal stores. We tested adenosine triphosphate (ATP) -mediated Ca2+ responses of DCs transiently exposed to nanomolar THI. Transcriptional and immunocytochemical analyses show that murine myeloid immature DCs (IDCs) and mature DCs (MDCs) express inositol 1,4,5-trisphosphate receptor (IP3R) and ryanodine receptor (RyR) Ca2+ channels, known targets of THI. IDCs express the RyR1 isoform in a punctate distribution that is densest near plasma membranes and within dendritic processes, whereas IP3Rs are more generally distributed. RyR1 positively and negatively regulates purinergic signaling because ryanodine (Ry) blockade a) recruited 80% more ATP responders, b) shortened ATP-mediated Ca2+ transients > 2-fold, and c) produced a delayed and persistent rise (>/= 2-fold) in baseline Ca2+. THI (100 nM, 5 min) recruited more ATP responders, shortened the ATP-mediated Ca2+ transient (>/= 1.4-fold) , and produced a delayed rise (>/= 3-fold) in the Ca2+ baseline, mimicking Ry. THI and Ry, in combination, produced additive effects leading to uncoupling of IP3R and RyR1 signals. THI altered ATP-mediated interleukin-6 secretion, initially enhancing the rate of cytokine secretion but suppressing cytokine secretion overall in DCs.DCs are exquisitely sensitive to THI, with one mechanism involving the uncoupling of positive and negative regulation of Ca2+ signals contributed by RyR1.
Collapse
Affiliation(s)
- Samuel R Goth
- National Institute of Environmental Health Sciences Center for Children's Environmental Health, and Department of Veterinary Molecular Biosciences, University of California-Davis, Davis, California, USA
| | | | | | | | | |
Collapse
|
24
|
Yang YJ, Wang ZY, Chen SH, Ge XR. Cloning and characterization of a new isoform of mouse interleukin-18. Acta Biochim Biophys Sin (Shanghai) 2005; 37:826-34. [PMID: 16331327 DOI: 10.1111/j.1745-7270.2005.00116.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Interleukin-18 (IL-18) is a novel proinflammatory cytokine with potent interferon (IFN)-lambda inducing activity that plays an important biological role in the enhancement of the activity of natural killer cells and cytotoxic T lymphocytes. In this study, we have identified a novel short form of IL-18 in mouse, named IL-18s. IL-18s might be an alternative splicing variant of IL-18 and its cDNA contains a 57 bp in-frame deletion. Like IL-18, IL-18s is also widely expressed in mouse tissues. It was suggested that IL-18s might have a caspase-1-dependent mechanism for maturation and secretion similar to that of IL-18: when transfected in COS-7 cells, pro-IL-18s (22 kDa) could be detected, and the mature IL-18s (16 kDa) could also be detected when combined with caspase-1. We observed that recombinant mouse IL-18s did not show any IL-18-like function, and IL-18s could enhance the ability of IL-18 to increase IFN-lambda production by approximately 40% in mouse splenocytes. This effect was observed primarily at relative low concentrations of IL-18, suggesting that IL-18s might regulate the activity of IL-18 in the physiological conditions.
Collapse
Affiliation(s)
- Yong-Jie Yang
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences; Graduate School of the Chinese Academy of Sciences, Shanghai 200031 China
| | | | | | | |
Collapse
|
25
|
O'Connell PJ, Wang X, Leon-Ponte M, Griffiths C, Pingle SC, Ahern GP. A novel form of immune signaling revealed by transmission of the inflammatory mediator serotonin between dendritic cells and T cells. Blood 2005; 107:1010-7. [PMID: 16223770 PMCID: PMC1895901 DOI: 10.1182/blood-2005-07-2903] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Adaptive immunity is triggered at the immune synapse, where peptide-major histocompatibility complexes and costimulatory molecules expressed by dendritic cells (DCs) are physically presented to T cells. Here we describe transmission of the inflammatory monoamine serotonin (5-hydroxytryptamine [5-HT]) between these cells. DCs take up 5-HT from the microenvironment and from activated T cells (that synthesize 5-HT) and this uptake is inhibited by the antidepressant, fluoxetine. Expression of 5-HT transporters (SERTs) is regulated by DC maturation, exposure to microbial stimuli, and physical interactions with T cells. Significantly, 5-HT sequestered by DCs is stored within LAMP-1+ vesicles and subsequently released via Ca2+-dependent exocytosis, which was confirmed by amperometric recordings. In turn, extracellular 5-HT can reduce T-cell levels of cAMP, a modulator of T-cell activation. Thus, through the uptake of 5-HT at sites of inflammation, and from activated T cells, DCs may shuttle 5-HT to naive T cells and thereby modulate T-cell proliferation and differentiation. These data constitute the first direct measurement of triggered exocytosis by DCs and reveal a new and rapid type of signaling that may be optimized by the intimate synaptic environment between DCs and T cells. Moreover, these results highlight an important role for 5-HT signaling in immune function and the potential consequences of commonly used drugs that target 5-HT uptake and release.
Collapse
|
26
|
Strand ML, Wahlgren A, Svechnikov K, Zetterström C, Setchell BP, Söder O. Interleukin-18 is expressed in rat testis and may promote germ cell growth. Mol Cell Endocrinol 2005; 240:64-73. [PMID: 16002206 DOI: 10.1016/j.mce.2005.05.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Revised: 05/16/2005] [Accepted: 05/25/2005] [Indexed: 10/25/2022]
Abstract
Although host-defence mechanisms, designed to preserve the integrity of the developing germ cells are operative in the testis, the components of this protective system have yet to be characterised in detail. Here, we report that the cytokine interleukin-18 (IL-18) is expressed in the rat testis and may contribute to these defences. Thus, analysis by RT-PCR and Western blotting revealed pronounced testicular expression of pro-IL-18 from postnatal day 5 and onwards. Expression of both IL-18 mRNA and protein was found to be localised to meiotic and post-meiotic germ cells as evaluated by in situ hybridisation and immunohistochemistry, respectively. The mRNA species coding for the IL-18 receptor and IL-1beta converting enzyme, which activates pro-IL-18, were also shown to be expressed by the seminiferous tubules. Recombinant IL-18 was seen to stimulate spermatogonial DNA synthesis in cultures of staged segments of rat seminiferous tubules, without influencing germ cell apoptosis. These results suggest that IL-18 may have host-protective and growth-promoting functions in the testis, but further investigations need to be done to confirm this.
Collapse
Affiliation(s)
- Mona-Lisa Strand
- Department of Woman and Child Health, Paediatric Endocrinology Unit Q2:08, Astrid Lindgren Children's Hospital, Karolinska Institute and University Hospital, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
27
|
Semino C, Angelini G, Poggi A, Rubartelli A. NK/iDC interaction results in IL-18 secretion by DCs at the synaptic cleft followed by NK cell activation and release of the DC maturation factor HMGB1. Blood 2005; 106:609-616. [PMID: 15802534 DOI: 10.1182/blood-2004-10-3906] [Citation(s) in RCA: 260] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interaction of natural killer (NK) cells with autologous immature dendritic cells (DCs) results in reciprocal activation; however, the underlying mechanisms are so far elusive. We show here that NK cells trigger immature DCs to polarize and secrete interleukin 18 (IL-18), a cytokine lacking a secretory leader sequence. This occurs through a Ca2+-dependent and tubulin-mediated recruitment of IL-18-containing secretory lysosomes toward the adhering NK cell. Lysosome exocytosis and IL-18 secretion are restricted at the synaptic cleft, thus allowing activation of the interacting NK cells without spreading of the cytokine. In turn, DC-activated NK cells secrete the proinflammatory cytokine high mobility group B1 (HMGB1), which induces DC maturation and protects DCs from lysis. Also HMGB1 is a leaderless cytokine that undergoes regulated secretion. Differently from IL-18, soluble HMGB1 is consistently detected in NK/DC supernatants. These data point to secretion of leaderless cytokines as a key event for the reciprocal activation of NK cells and DCs. DCs initiate NK cell activation by targeted delivery of IL-18, thus instructing NK cells in the absence of adaptive-type cytokines; in turn, activated NK cells release HMGB1, which promotes inflammation and induces DC maturation, thus favoring the onset of the adaptive immune response.
Collapse
Affiliation(s)
- Claudia Semino
- Laboratory of Cell Biology, Department of Translational Oncology, National Institute for Cancer Research, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | | | | | | |
Collapse
|
28
|
DeMarco RA, Fink MP, Lotze MT. Monocytes promote natural killer cell interferon gamma production in response to the endogenous danger signal HMGB1. Mol Immunol 2005; 42:433-44. [PMID: 15607795 DOI: 10.1016/j.molimm.2004.07.023] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Substantial attention has been paid to the role of the toll-like receptor (TLR) ligands of late and their role in regulating the innate immune response. They serve as exogenous danger signals important in informing and driving the distal adaptive immune response to pathogens. Less clear has been the role of the nominal endogenous danger signals released and recognized in stressed cells following genotoxic or metabolic stress as occurs in progressively growing tumors. HMGB1 (high-mobility group B1) is a nuclear protein well characterized for its ability to modify DNA access to transcriptional proteins that is released from necrotic cells as well as secreted through the endosomal route from hematopoietic cells, serving as a late mediator of sepsis. It interacts with high-affinity RAGE (receptor for advanced glycation end products) and TLR2 receptors. Here we show that HMGB1 enhances interferon gamma release from macrophage (but not dendritic cell)-stimulated NK cells. This is effective only when coupled with other pro-inflammatory cytokines particularly with IL-2 in combination with IL-1 or IL-12. We have used this information to suggest that HMGB1, which also promotes epithelial migration and proliferation, drives repair in the absence or inhibition of other factors but enhances inflammation in their presence. The implications for tumorigenesis and tumor progression are quite important as they may be for other states of chronic inflammation.
Collapse
Affiliation(s)
- Richard A DeMarco
- Department of Surgery, Molecular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | | | | |
Collapse
|
29
|
Bufler P, Gamboni-Robertson F, Azam T, Kim SH, Dinarello CA. Interleukin-1 homologues IL-1F7b and IL-18 contain functional mRNA instability elements within the coding region responsive to lipopolysaccharide. Biochem J 2004; 381:503-10. [PMID: 15046617 PMCID: PMC1133858 DOI: 10.1042/bj20040217] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2004] [Revised: 03/25/2004] [Accepted: 03/26/2004] [Indexed: 12/16/2022]
Abstract
IL-1F7b, a novel homologue of the IL-1 (interleukin 1) family, was discovered by computational cloning. We demonstrated that IL-1F7b shares critical amino acid residues with IL-18 and binds to the IL-18-binding protein enhancing its ability to inhibit IL-18-induced interferon-gamma. We also showed that low levels of IL-1F7b are constitutively present intracellularly in human blood monocytes. In this study, we demonstrate that similar to IL-18, both mRNA and intracellular protein expression of IL-1F7b are up-regulated by LPS (lipopolysaccharide) in human monocytes. In stable transfectants of murine RAW264.7 macrophage cells, there was no IL-1F7b protein expression despite a highly active CMV promoter. We found that IL-1F7b-specific mRNA was rapidly degraded in transfected cells, via a 3'-UTR (untranslated region)-independent control of IL-1F7b transcript stability. After LPS stimulation, there was a rapid transient increase in IL-1F7b-specific mRNA and concomitant protein levels. Using sequence alignment, we found a conserved ten-nucleotide homology box within the open reading frame of IL-F7b, which is flanking the coding region instability elements of some selective genes. In-frame deletion of downstream exon 5 from the full-length IL-1F7b cDNA markedly increased the levels of IL-1F7b mRNA. A similar coding region element is located in IL-18. When transfected into RAW264.7 macrophages, IL-18 mRNA was also unstable unless treated with LPS. These results indicate that both IL-1F7b and IL-18 mRNA contain functional instability determinants within their coding region, which influence mRNA decay as a novel mechanism to regulate the expression of IL-1 family members.
Collapse
Affiliation(s)
- Philip Bufler
- Division of Infectious Diseases, University of Colorado Health Sciences Center, 4200 East Ninth Ave., Denver, CO 80262, USA.
| | | | | | | | | |
Collapse
|
30
|
Gaggero A, De Ambrosis A, Mezzanzanica D, Piazza T, Rubartelli A, Figini M, Canevari S, Ferrini S. A novel isoform of pro-interleukin-18 expressed in ovarian tumors is resistant to caspase-1 and -4 processing. Oncogene 2004; 23:7552-60. [PMID: 15326478 DOI: 10.1038/sj.onc.1208036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interleukin-18 (IL-18) is a proinflammatory cytokine synthesized as a 24 kDa inactive precursor (pro-IL-18) by several cell types, and is processed to a bioactive molecule of 18 kDa by the proteinases caspase-1 or caspase-4. All ovarian carcinoma cell lines express pro-IL-18, only in some instances coexpress caspase-1, and always express caspase-4; in any case, they display a defective processing of IL-18. We analysed whether pro-IL-18, present in two ovarian carcinoma cell lysates, could be processed 'in vitro' by recombinant active caspase-1. While most of pro-IL-18 could be cleaved by caspase-1, a residual of pro-IL-18 appeared to be resistant. Cloning and sequence analysis of the whole pro-IL-18 open reading frame demonstrated the existence of an alternatively spliced mRNA variant, which lacked exon-3 (Delta3pro-IL-18). The 12 bp exon-3 encodes for the AEDD amino-acid sequence, which is N-terminal with respect to the cleavage site of caspase-1. Both pro-IL-18 and Delta3pro-IL-18 mRNA isoforms were detected in all ovarian cancer cell lines analysed, while Delta3pro-IL-18 mRNA was undetectable in normal ovarian epithelial cells. The Delta3pro-IL-18 cDNA induced synthesis of an alternative Delta3pro-IL-18 protein upon transfection into a murine cell line. The Delta3pro-IL-18 protein was resistant to proteolytic activation by caspase-1 and -4, although it was capable to bind caspase-1. Aternative splicing of pro-IL-18 exon-3 may represent a novel mechanism of regulation of bioactive IL-18 production in human ovarian tumors.
Collapse
Affiliation(s)
- Alessia Gaggero
- Istituto Nazionale per la Ricerca sul Cancro, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Interleukin (IL)-18 is a new member of the IL-1 family of proinflammatory cytokines. Based on preclinical studies in animals, IL-18 likely plays a role in rheumatoid arthritis, and strategies to block IL-18 activity are underway in clinical trials. In one of these trials,a naturally occurring IL-18 binding protein (IL-18 BP) binds IL-18 with a high affinity and reduces disease severity in models of inflammatory diseases. IL-18 BP is not the soluble receptor for IL-18 but rather a distinct molecule, which appears to be distantly related to the IL-1 receptor type II, both structurally and functionally, and hence represents part of the IL-1 family of receptors.
Collapse
Affiliation(s)
- Charles A Dinarello
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, 4200 East Ninth Avenue, B168, Denver, CO 80262, USA
| |
Collapse
|
32
|
Dinarello CA, Novick D, Rubinstein M, Lonnemann G. Interleukin 18 and interleukin 18 binding protein: possible role in immunosuppression of chronic renal failure. Blood Purif 2004; 21:258-70. [PMID: 12784053 DOI: 10.1159/000070699] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although interleukin (IL)-18 is a member of the IL-1 family of ligands, IL-18 appears to have unique characteristics, particularly in the regulation of the T helper type 1 (Th1) response. Th1 responses are required for tumor surveillance, killing intracellular organisms, and to provide help for antibody production. In patients with chronic renal failure, the well-known immunosuppression contributes to a failure to respond to infectious challenges and vaccinations. The most salient biological property of IL-18, linking this cytokine to the Th1 response, is its ability to induce interferon gamma (IFN-gamma). In fact, IL-18 was originally identified as an IFN-gamma-inducing factor, and IFN-gamma production is the hallmark of the Th1 response. Dysregulation of IFN-gamma production resulting from reduced activity of IL-18 would explain one of the mechanisms of immunosuppression in patients with chronic renal failure. The activity of IL-18 can be regulated by the IL-18-binding protein (IL-18BP), a glycoprotein of 40,000 daltons, which is constitutively expressed and appears to be the natural inhibitor of IL-18 activity. Unlike soluble receptors for IL-18, IL-18BP does not have a transmembrane domain; IL-18BP is a secreted protein possessing a high-affinity binding and ability to neutralize IL-18. IL-18BP was discovered in human urine and is excreted in health following glomerular filtration. With decreasing renal function, the concentrations of IL-18BP in the circulation are elevated as compared with subjects with a normal renal function, and these elevated levels may result in a decreased IL-18 activity. Because of the importance of IL-18 and IFN-gamma in the Th1 response, the biology of IL-18 and IL-18BP is reviewed here in the context of the immunosuppression of chronic renal failure.
Collapse
Affiliation(s)
- Charles A Dinarello
- Department of Medicine, University of Colorado Health Sciences Center, Denver, Colo 80262, USA
| | | | | | | |
Collapse
|
33
|
Bonaldi T, Talamo F, Scaffidi P, Ferrera D, Porto A, Bachi A, Rubartelli A, Agresti A, Bianchi ME. Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. EMBO J 2003; 22:5551-60. [PMID: 14532127 PMCID: PMC213771 DOI: 10.1093/emboj/cdg516] [Citation(s) in RCA: 1017] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2003] [Revised: 08/11/2003] [Accepted: 08/15/2003] [Indexed: 12/29/2022] Open
Abstract
High Mobility Group 1 protein (HMGB1) is a chromatin component that, when leaked out by necrotic cells, triggers inflammation. HMGB1 can also be secreted by activated monocytes and macrophages, and functions as a late mediator of inflammation. Secretion of a nuclear protein requires a tightly controlled relocation program. We show here that in all cells HMGB1 shuttles actively between the nucleus and cytoplasm. Monocytes and macrophages acetylate HMGB1 extensively upon activation with lipopolysaccharide; moreover, forced hyperacetylation of HMGB1 in resting macrophages causes its relocalization to the cytosol. Cytosolic HMGB1 is then concentrated by default into secretory lysosomes, and secreted when monocytic cells receive an appropriate second signal.
Collapse
Affiliation(s)
- Tiziana Bonaldi
- DIBIT, San Raffaele Scientific Institute, San Raffaele University, via Olgettina 58, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Gudipaty L, Munetz J, Verhoef PA, Dubyak GR. Essential role for Ca2+ in regulation of IL-1beta secretion by P2X7 nucleotide receptor in monocytes, macrophages, and HEK-293 cells. Am J Physiol Cell Physiol 2003; 285:C286-99. [PMID: 12660148 DOI: 10.1152/ajpcell.00070.2003] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Interleukin (IL)-1beta is a proinflammatory cytokine that elicits the majority of its biological activity extracellularly, but the lack of a secretory signal sequence prevents its export via classic secretory pathways. Efficient externalization of IL-1beta in macrophages and monocytes can occur via stimulation of P2X7 nucleotide receptors with extracellular ATP. However, the exact mechanisms by which the activation of these nonselective cation channels facilitates secretion of IL-1beta remain unclear. Here we demonstrate a pivotal role for a sustained increase in cytosolic Ca2+ to potentiate secretion of IL-1beta via the P2X7 receptors. Using HEK-293 cells engineered to coexpress P2X7 receptors with mature IL-1beta (mIL-1beta), we show that activation of P2X7 receptors results in a rapid secretion of mIL-1beta by a process(es) that is dependent on influx of extracellular Ca2+ and a sustained rise in cytosolic Ca2+. Moreover, reduction in extracellular Ca2+ attenuates approximately 90% of P2X7 receptor-mediated IL-1beta secretion but has no effect on enzymatic processing of precursor IL-1beta (proIL-1beta) to mIL-1beta by caspase-1. Similar experiments with THP-1 human monocytes and Bac1.2F5 murine macrophages confirm the unique role of Ca2+ in P2X7 receptor-mediated secretion of IL-1beta. In addition, we report that cell surface expression of P2X7 receptors in the absence of external stimulation also results in enhanced release of IL-1beta and that this can be repressed by inhibitors of P2X7 receptors. We clarify an essential role for Ca2+ in ATP-induced IL-1beta secretion and indicate an additional role of P2X7 receptors as enhancers of the secretory apparatus by which IL-1beta is released.
Collapse
Affiliation(s)
- Lalitha Gudipaty
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
35
|
Dinarello CA, Fantuzzi G. Interleukin-18 and host defense against infection. J Infect Dis 2003; 187 Suppl 2:S370-84. [PMID: 12792854 DOI: 10.1086/374751] [Citation(s) in RCA: 216] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Interferon (IFN)-gamma-inducing factor was previously termed interleukin (IL)-18. Although IL-12 is also an IFN-gamma-inducing factor, the activity of IL-18 (but not IL-12) in models of sepsis and death is dependent on the intracellular cysteine protease IL-1beta converting enzyme (caspase-1). Caspase-1 is required for cleavage of the inactive precursor form of IL-18 into an active cytokine, and caspase-1-deficient mice are resistant to lethal endotoxemia. The absence of IFN-gamma (but not IL-1beta) in caspase-1-deficient mice is responsible for this resistance. However, the role of IFN-gamma in murine defense against gram-negative infection is inconsistent. Mice deficient in IFN-gamma are not resistant to lethal endotoxemia but are resistant when treated with neutralizing antibodies to IL-18 and challenged with a lethal injection of some endotoxins. Anti-IL-18 treatment also reduces neutrophil accumulation in liver and lungs. Neutralizing IL-18 with the IL-18 binding protein protects mice against endotoxin- and ischemia-induced hepatic damage. Thus, blockade of IL-18 appears to be a viable clinical target to combat the pathologic consequences of sepsis via IFN-gamma mechanisms.
Collapse
Affiliation(s)
- Charles A Dinarello
- Department of Medicine, Division of Infectious Diseases, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | |
Collapse
|
36
|
Eaton AD, Xu D, Garside P. Administration of exogenous interleukin-18 and interleukin-12 prevents the induction of oral tolerance. Immunology 2003; 108:196-203. [PMID: 12562328 PMCID: PMC1782890 DOI: 10.1046/j.1365-2567.2003.01570.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2002] [Revised: 10/14/2002] [Accepted: 10/24/2002] [Indexed: 01/31/2023] Open
Abstract
Interleukin-18 (IL-18), a pro-inflammatory member of the IL-1 family, has been associated with autoimmunity and allergic disease. This type of pathology is thought to be the result of a defect in immunological tolerance and is often observed in inflammatory disorders of the gut such as coeliac disease, Crohn's disease and ulcerative colitis. IL-18 has been implicated in a number of mucosal immune disorders, where it synergizes with IL-12 to induce the production of interferon-gamma (IFN-gamma). We have therefore investigated the effects of IL-18 and IL-12 administration on the induction of oral tolerance to ovalbumin. The suppression of specific Ig G2a production, delayed-type hypersensitivity responses and IFN-gamma production by antigen-specific T cells were all abrogated by the presence of exogenous IL-12 and IL-18, suggesting that oral tolerance was broken. The expression of the co-stimulatory molecule CD80 on dendritic cells was also shown to be increased by this combination of cytokines. As dendritic cells are thought to be of major importance in the induction of tolerance, this suggests a mechanism by which tolerance to mucosal antigens may be broken in vivo.
Collapse
Affiliation(s)
- Alfred D Eaton
- Division of Immunology, Infection & Inflammation, University of Glasgow, Western Infirmary, Glasgow, UK
| | | | | |
Collapse
|
37
|
Dreher D, Kok M, Obregon C, Kiama SG, Gehr P, Nicod LP. Salmonella
virulence factor SipB induces activation and release of IL‐18 in human dendritic cells. J Leukoc Biol 2002. [DOI: 10.1189/jlb.72.4.743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Donatus Dreher
- Division of Pneumology, University Hospital of Geneva, Switzerland
| | - Menno Kok
- Department of Genetics and Microbiology and Department of Medical Biochemistry, University of Geneva, Switzerland
| | - Carolina Obregon
- Division of Pneumology, University Hospital of Geneva, Switzerland
| | - Stephen G. Kiama
- Institute of Anatomy, University of Berne, Switzerland; and
- Department of Veterinary Anatomy, University of Nairobi, Kenya
| | - Peter Gehr
- Institute of Anatomy, University of Berne, Switzerland; and
| | - Laurent P. Nicod
- Division of Pneumology, University Hospital of Geneva, Switzerland
| |
Collapse
|
38
|
Weber W, Fussenegger M. Artificial mammalian gene regulation networks-novel approaches for gene therapy and bioengineering. J Biotechnol 2002; 98:161-87. [PMID: 12141985 DOI: 10.1016/s0168-1656(02)00130-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Recently developed strategies for targeted molecular interventions in mammalian cells have created novel opportunities in biotechnological and biomedical research with huge economic and therapeutic impact: the design of mammalian cells with desired phenotypes for biopharmaceutical manufacturing, tissue engineering and gene therapy. These advances have been enabled by constructing artificial gene regulation systems with control modalities similar to those evolved in key regulatory networks of mammalian cells. This review highlights recurring cellular regulation strategies and artificial gene regulation technology currently in use for rational reprogramming of cellular key events including metabolism, growth, differentiation and cell death to achieve sophisticated bioprocess and therapeutic goals.
Collapse
Affiliation(s)
- Wilfried Weber
- Institute of Biotechnology, Swiss Federal Institute of Technology, ETH Zurich ETH Hoenggerberg, HPT, Switzerland
| | | |
Collapse
|
39
|
Wang ZY, Gaggero A, Rubartelli A, Rosso O, Miotti S, Mezzanzanica D, Canevari S, Ferrini S. Expression of interleukin-18 in human ovarian carcinoma and normal ovarian epithelium: evidence for defective processing in tumor cells. Int J Cancer 2002; 98:873-8. [PMID: 11948465 DOI: 10.1002/ijc.10268] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Interleukin-18 (IL-18) is a proinflammatory monokine structurally related to IL-1beta that stimulates interferon-gamma (IFN-gamma) production. IL-18 is synthesized as an inactive precursor, pro-IL-18, which is cleaved by IL-1beta-converting enzyme (ICE)/caspase-1 in a mature protein. In view of the proposed use of IL-18 in cancer immuno/gene therapy, we have studied the expression of IL-18 in tumor cells. IL-18 mRNA was detected by reverse transcriptase polymerase chain reaction in all human ovarian carcinoma cell lines tested (9/9) and in one-half of tumor cell populations obtained from ovarian carcinoma patients (4/8). ICE mRNA was expressed in a smaller fraction of samples (3/9 cell lines and 3/8 samples from patients). IL-18 protein was also found in 7/13 ovarian carcinoma solid tumors by immunohistochemic analysis. In tumor cell lines we were able to detect abundant intracellular pro-IL-18 (24 kDa) by Western blotting, whereas the mature form of IL-18 was undetectable, irrespective of the presence of ICE mRNA and protein. Only pro-IL-18 was also found in the ovarian carcinoma cell supernatants, which did not display any IL-18 biologic activity in functional assays. Normal cultured ovarian epithelial cells revealed the presence of both IL-18 and ICE mRNA in all samples (5/5) and IL-18 protein was expressed by the thin epithelial cell layer surrounding normal ovary. More importantly, normal ovarian epithelial cells released low but detectable amounts of mature IL-18 in the culture supernatant, which displayed IL-18-like biologic activity in functional assays. These data suggest that mature biologically active IL-18 production is a feature of the normal ovarian surface epithelium lost during neoplastic transformation.
Collapse
Affiliation(s)
- Zhao Yuan Wang
- Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Angelini G, Gardella S, Ardy M, Ciriolo MR, Filomeni G, Di Trapani G, Clarke F, Sitia R, Rubartelli A. Antigen-presenting dendritic cells provide the reducing extracellular microenvironment required for T lymphocyte activation. Proc Natl Acad Sci U S A 2002; 99:1491-6. [PMID: 11792859 PMCID: PMC122218 DOI: 10.1073/pnas.022630299] [Citation(s) in RCA: 296] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
T lymphocytes are defective in cystine uptake and thus require exogenous thiols for activation and function. Here we show that monocyte-derived human dendritic cells (DCs) release cysteine in the extracellular space. Cysteine generation is increased by lipopolysaccharide and tumor necrosis factor alpha, and by contact with T cells specifically recognizing soluble or alloantigens. These stimuli also induce thioredoxin (TRX) accumulation in DCs. However, only the contact with antigen-specific T cells triggers TRX secretion by the antigen-presenting cells. Fewer extracellular thiols are recovered after DC-T cell interactions when cystine uptake or TRX activity are inhibited. In addition, glutamate (Glu) and anti-TRX-inactivating antibodies inhibit antigen-dependent T lymphocyte proliferation. These findings indicate that, during antigen presentation, DCs uptake cystine and release cysteine and TRX, thus providing a reducing microenvironment that facilitates immune response.
Collapse
Affiliation(s)
- Giovanna Angelini
- Protein Biology Unit, National Cancer Research Institute, 16132 Genoa, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Gardella S, Andrei C, Lotti LV, Poggi A, Torrisi MR, Zocchi MR, Rubartelli A. CD8(+) T lymphocytes induce polarized exocytosis of secretory lysosomes by dendritic cells with release of interleukin-1beta and cathepsin D. Blood 2001; 98:2152-2159. [PMID: 11568002 DOI: 10.1182/blood.v98.7.2152] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We recently reported that human dendritic cells release the leaderless secretory protein interleukin-1beta (IL-1beta) following specific interaction with alloreactive T lymphocytes. To clarify the molecular mechanism underlying this secretion, this study investigated the intracellular trafficking of IL-1beta in dendritic cells and the signal(s) regulating its release. Results show that a fraction of the intracellular IL-1beta precursor colocalizes with the hydrolase cathepsin D in endolysosomes of dendritic cells; secretion of both proteins is elicited by stimuli that induce intracellular calcium increases. Alloreactive CD8(+) T lymphocytes generate a Ca(++) influx in dendritic cells followed by enrichment in endolysosomes containing IL-1beta and cathepsin D beneath the membrane in contact with T cells. These events result in polarized exocytosis of secretory lysosomes, mediated by microtubules, with release of IL-1beta and cathepsin D toward the interacting CD8(+) T cell.
Collapse
Affiliation(s)
- S Gardella
- Unit of Protein Biology, Laboratory of Immunology, and Biotechnology Section of Roma, National Cancer Research Institute, 16132 Genoa, Italy
| | | | | | | | | | | | | |
Collapse
|
42
|
Caron G, Delneste Y, Roelandts E, Duez C, Herbault N, Magistrelli G, Bonnefoy JY, Pestel J, Jeannin P. Histamine induces CD86 expression and chemokine production by human immature dendritic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:6000-6. [PMID: 11342615 DOI: 10.4049/jimmunol.166.10.6000] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mast cells and immature dendritic cells (DC) are in close contact in peripheral tissues. Upon activation, mast cells release histamine, a mediator involved in the immediate hypersensitivity reaction. We therefore tested whether histamine could affect human DC activation and maturation. Histamine induces CD86 expression on immature DC in a dose-dependent (significant at 10(-7) M) and transient manner (maximal after 24-h stimulation). Histamine also transiently up-regulates the expression of the costimulatory and accessory molecules, CD40, CD49d, CD54, CD80, and MHC class II. As a consequence, immature DC exposed for 24 h to histamine stimulate memory T cells more efficiently than untreated DC. In addition, histamine induces a potent production of IL-6, IL-8, monocyte chemoattractant protein 1, and macrophage-inflammatory protein 1alpha by immature DC and also up-regulates IL-1beta, RANTES, and macrophage-inflammatory protein 1beta but not TNF-alpha and IL-12 mRNA expression. Histamine activates immature DC through both the H1 and H2 receptors. However, histamine-treated DC do not have a phenotype of fully mature cells, as they do neither show significant changes in the expression of the chemokine receptors, CCR5, CCR7 and CXC chemokine receptor 4, nor expression of CD83 de novo. These data demonstrate that histamine activates immature DC and induces chemokine production, thereby suggesting that histamine, via stimulation of resident DC, may participate locally in T cell stimulation and in the late inflammatory reaction associated with allergic disorders.
Collapse
Affiliation(s)
- G Caron
- Centre d'Immunologie Pierre Fabre, Saint-Julien en Genevois, France
| | | | | | | | | | | | | | | | | |
Collapse
|