Skip to main content
Erschienen in: Immunologic Research 3/2015

Open Access 01.03.2015

Roles of retinoic acid-inducible gene-I-like receptors (RLRs), Toll-like receptor (TLR) 3 and 2′-5′ oligoadenylate synthetase as viral recognition receptors on human mast cells in response to viral infection

verfasst von: Mizuho Tsutsui-Takeuchi, Hiroko Ushio, Minoru Fukuda, Takahiko Yamada, François Niyonsaba, Ko Okumura, Hideoki Ogawa, Shigaku Ikeda

Erschienen in: Immunologic Research | Ausgabe 3/2015

Abstract

To investigate the anti-viral responses of human mast cells, we performed PCR array analysis of these cells after infection with vesicular stomatitis virus (VSV). PCR array analysis revealed that human mast cells up-regulated several anti-viral genes, including melanoma differentiation-associated gene 5, retinoic acid-inducible gene-I, and Toll-like receptor 3, together with type I interferons and chemokines, upon VSV infection. Additionally, we found that 2′-5′ oligoadenylate synthetase, which also works as a virus recognition receptor by activating the latent form of RNase L, leading to viral RNA degradation, was up-regulated in human mast cells upon VSV infection. Moreover, small interfering RNA analysis to identify the receptors responsible for mast cell activation by VSV revealed that these receptors reciprocally cooperate to produce anti-viral cytokines and chemokines, inhibiting VSV replication. Our findings suggest that human mast cells produce cytokines and chemokines using several viral recognition receptors, leading to the inhibition of viral replication. These data provide novel information that improves our understanding of the roles of human mast cells in immune responses against viruses.

Introduction

Mast cells are tissue resident cells derived from hematopoietic precursor cells [1]. Mast cells reside in the tissues closely associated with blood vessels or on the surface of the body, particularly in the skin and mucosal membranes. Thus, in addition to their roles as effector cells in IgE-mediated allergic diseases, mast cells are widely considered to be important innate immune cells [2, 3].
The roles of mast cells in immune surveillance and innate immunity against bacterial pathogens have been well defined [4]. Generally, host innate immune responses against various pathogens are initiated by the recognition of microbial-specific components, such as lipopolysaccharides (LPSs), lipoproteins, flagellins and nucleic acids, by pattern recognition receptors (PRRs). The recognition of microbial components by PRRs results in the coordinated activation of transcription factors, leading to the expression of inflammatory cytokines, chemokines and type I interferons (IFNs). We and others have previously shown that mast cells contribute to innate immune responses against invading bacteria by expressing PRRs, such as TLRs-2, 3, 4, 5, 6, 7 and 9, which respond to specific ligands by inducing the production of cytokines and chemokines or the release of the cell’s granular contents [57]. Similarly, innate immune responses against viral infection begin with the recognition of the virus by specific PRRs. Viral nucleic acids are known to be recognized by distinct types of sensors: TLRs, which detect double-stranded (ds) or single-stranded (ss) RNA in the endosome; retinoic acid-inducible gene-I-like receptors (RLRs), which recognize viral RNA in the cytoplasm; and DNA sensors, which detect cytoplasmic viral DNA [8]. The activation of mast cells, including the specific production of anti-viral cytokines such as type I interferon or the release of their granular contents, has been observed upon stimulation with viruses, virus products or poly I:C [914]. Additionally, evidence regarding the roles of mast cells in response to viral infections has been accumulating. Orinska et al. [15] reported the functional consequences of mast cell activation in response to viral infection, demonstrating that mast cells stimulated via TLR3 produced chemokines that mediated CD8+ T cell recruitment in vivo. Burke et al. [16] demonstrated that poly I:C-exposed or reovirus-infected mast cells recruit NK cells in a CXCL8-dependent manner. Furthermore, the TLR-induced activation of mast cells by LPS or poly I:C enhanced the capacity of mast cells to activate CD8+ T cells [17]. More recently, mast cells have been shown to play important roles in host protection against herpes simplex virus 2 (HSV-2) infection [18]. Thus, clarifying the mechanisms of virus recognition that lead to mast cell activation is important for understanding the roles played by mast cells in virus infection because mast cells work not only as innate immune cells for host protection, but also as exaggerators of allergic diseases associated with virus infection.
Although the knowledge regarding how mast cells are activated by viruses or virus products is limited, we previously demonstrated that mouse mast cells express several recognition receptors for viral products and play important roles in early anti-viral cytokine responses via the recognition of viral products by these receptors [19]. Because the differences between human and rodent mast cells reside not only in their phenotypic differences but also in the expression of innate immune receptors such as PRRs and RLRs, in this study, we further analyzed the anti-viral responses of human mast cells using a PCR array system. We found that 2′-5′ oligoadenylate synthetase (OAS), which also functions as a virus recognition receptor by activating the latent form of RNase L, leading to viral RNA degradation, was also responsible for the activation of human mast cells by the virus, collaborating with RLRs and TLR3.

Materials and methods

Cells

The Laboratory of Allergic Diseases 2 cell line (LAD2), which was isolated from the bone marrow of a patient with mast cell leukemia, was a gift from Dr. Arnold Kirshenbaum (National Institutes of Health, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA). The LAD2 cells were cultured at 37 °C in StemPro-34 serum-free medium (Gibco by Life Technologies, Tokyo, Japan) supplemented with StemPro-34 nutrient (Gibco), l-glutamine (200 mM) (Gibco), penicillin (100 U/ml) and streptomycin (100 μg/ml) (Meiji Seika Pharma Co., Ltd, Tokyo, Japan), as well as recombinant human stem cell factor and cytokines (100 ng/ml) (PeproTech, Rocky Hill, NJ, USA).

Virus infection

Vesicular stomatitis virus (VSV) was kindly provided by Drs. K. Honda and H. Yanai (The University of Tokyo). LAD2 cells (1 × 106 cells/ml) were stimulated in medium without antibiotics with VSV at the indicated multiplicities of infection (MOIs) for 1 to 24 h at 37 °C. To analyze the degranulation of the cells, LAD2 cells were resuspended at 1 × 106 cells/ml in Tyrode’s buffer containing 0.1 % BSA and stimulated with VSV at the indicated MOIs. The release of β-hexosaminidase was measured as previously described [5]. Degranulation was expressed as the percent release of β-hexosaminidase [(OD: stimulated-un-stimulated/OD: total lysate-un-stimulated) × 100 %].

Quantitative PCR

Total RNA was extracted from the cells using an RNAspin Mini kit (GE Healthcare, Buckinghamshire, UK) and treated with DNase I (GE Healthcare) and an RNeasy Plus Mini kit (Qiagen, Hilden, Germany). Complementary DNA was synthesized using SuperScript™ II reverse transcriptase (Invitrogen, Tokyo, Japan) with random primers. Quantitative PCR was performed using the TaqMan method with an ABI 7500 System (Applied Biosystems, Piscataway, NJ, USA). The mRNA levels were normalized to β-actin gene expression and expressed as the fold-induction relative to uninfected cells.

PCR array analysis

The Human Antiviral Response RT2 Profiler PCR Array (Qiagen) was used to profile the expression of 84 key genes involved in the innate antiviral immune response according to the manufacturer’s instructions.

Measurement of cytokines and chemokines

LAD2 cells (1 × 106/ml) were stimulated with the indicated concentrations of virus for 24 h. The IFN-α and IFN-β concentrations in the supernatants were determined using ELISA kits according to the manufacturer’s instructions (PBL Assay Science, Piscataway, NJ, USA). The IL-6, CCL5 (RANTES), CXCL10 (IP10), CXCL11 and IL-15 concentrations in the supernatants were also determined using ELISA kits according to the manufacturer’s instructions (R&D Systems, Inc. Minneapolis, MN, USA).

Western blotting

LAD2 cells were untreated or infected with VSV (MOI = 100 or 300) for 24 h. After stopping the reaction by adding ice-cold PBS, sample buffer containing SDS was directly added to the pellets, followed by brief sonication. Then, the lysates were subjected to SDS-PAGE and transferred onto polyvinylidene difluoride membranes (Merck Millipore, Tokyo, Japan). The membranes were analyzed by immunoblotting with anti-MDA5, anti-RIG-I (both from Cell Signaling Technology, Danvers, MA, USA), anti-TLR3 (Acris Antibodies, San Diego, CA, USA), anti-OAS2 (OriGene Technologies, Inc., Rockville, MD, USA) or anti-β-actin antibody (BioLegend, San Diego, CA, USA), followed by the respective HRP-conjugated secondary anti-immunoglobulin antibodies (GE Healthcare). The membranes were developed using Luminata™ Forte Western HRP substrate (Merck Millipore).

Small interfering RNA (siRNA) treatment

LAD2 cells were transfected with control or specific siRNA for rig-I, mda5, tlr3, oas1, oas2 and oas3 (Applied Biosystems) using the transfecting system NEON (program 16). The individual gene silencing efficiency was determined by quantitative PCR 24 h after transfection, and then the cells were used for experiments.

Measurement of virus titer

LAD2 cells were infected with VSV (MOI = 10) 24 h after the corresponding siRNA transfection. The titers of VSV in the supernatant of LAD2 cells 24 h after VSV infection were determined by counting the plaque forming units (PFUs) using Vero cells. Briefly, serially diluted supernatant of the LAD2 cells was incubated with Vero cells for 1 h. After removing the supernatant, the Vero cells were covered with 3% methylcellulose and incubated for another 24 h before counting the formed plaques.

Statistical analysis

A nonparametric Mann–Whitney U test was used. Values of p < 0.05 were considered statistically significant.

Results

Anti-viral responsive gene analysis of human mast cells upon virus infection using quantitative PCR array

Because we previously demonstrated that murine mast cells can respond to virus infection to produce type I interferon and several chemokines by up-regulating the RLRs, quantitative PCR array analysis of the innate immune response against viruses was performed in LAD2 cells after VSV infection. We found that several anti-viral responsive genes, including cytokines, chemokines and RLRs, were up-regulated 24 h after VSV (MOI = 100) infection (Table 1). Among these genes, we first identified the up-regulation of OAS genes that function as intracellular dsRNA sensors in human mast cells by activating the synthesis of 2′-5′-linked oligoadenylates (2-5As). Then, 2-5As activate the latent forms of RNase L, which leads to viral RNA degradation, thus acting as an anti-viral mechanism.
Table 1
Anti-viral responsive gene analysis of human mast cells upon virus infection using quantitative PCR array
Symbol
Fold change
CCL5
2.1425
CXCL10
1,360.225
CXCL11
509.8465
DDX58 (RIG-I)
14.1962
IFIH1 (MDA5)
9.1723
IFNA1
312.5437
IFNB1
623.459
IL15
3.2159
OAS2
52.4045
TLR3
370.1766
LAD2 cells were stimulated with the indicated concentrations of VSV (MOI = 100) for 24 h. The Human Antiviral Response RT2 Profiler PCR Array was used to profile the expression of 84 key genes involved in the innate antiviral immune response according to the manufacturer’s instructions

Viral infection induced anti-viral cytokine and chemokine responses in human mast cells

To confirm the PCR array results, first, we cultured LAD2 cells with different doses of VSV for 1–48 h. VSV (MOI = 1–100) infection resulted in both IFN-α and IFN-β induction in mast cells (Fig. 1a), and the peak time of the induction was approximately 16 h after infection (Fig. 1b). We also confirmed anti-viral chemokine expression by LAD2 cells upon viral infection. We observed CCL5 (RANTES), CXCL10 (IP-10), CXCL11, and IL-15 induction in LAD2 cells upon VSV infection (MOI = 1–100) (Fig. 2a). The time course analysis revealed that the peak expression times of CXCL10 and CXCL11 were similar to type I IFNs; in contrast, the induction of CCL5 and IL-15 seemed to be late compared with that of the other chemokines (Fig. 2b). Except for IL-15, the production of these cytokines and chemokines was also detectable at the protein level when measured by ELISA (Fig. 3).

VSV infection did not induce human mast cell degranulation

Because mast cell activation is often associated with degranulation, we examined whether VSV infection leads to mast cell degranulation. We could not detect significant β-hexosaminidase release after 1–24 h of VSV infection (Fig. 4). This result was not consistent with that obtained in murine mast cells, where we observed slight but significant β-hexosaminidase release from murine mast cells at 6 h, but not at 1 h, after VSV infection [19].

LAD2 cells expressed several RLRs and OAS that were up-regulated by VSV infection

Because we found that several intracellular receptors against dsRNA, including RLRs and OAS, were up-regulated in LAD2 cells upon VSV infection, we further examined whether these receptors play roles in the recognition and activation of human mast cells by VSV. First, we measured the mRNA levels of MDA5, RIG-I, TLR3, OAS1, OAS2, and OAS3 in un-stimulated mast cells by real-time PCR. TLR3 expression was overwhelmingly low compared with that of the other receptors (Fig. 5a). The expression of all of the receptors was markedly increased by VSV infection, with a significant difference (MOI = 100) after 24 h (Fig. 5b). The four OAS genes [OAS1, 2, 3 and OAS-like (OASL)] were identified in humans and have been mapped to chromosome 12. Among these genes, OAS1–3 have considerable homology to each other. The expression of OAS1 and OAS3 was abundant in un-stimulated mast cells (Fig. 5c). Although the expression of OAS2 was low in un-stimulated mast cells compared with OAS1 and 3, its expression level was drastically up-regulated upon VSV infection (Fig. 5d). We also analyzed the expression of MDA5, RIG-I, TLR3 and OAS2 at the protein level by Western blotting. Consistent with the mRNA results, increased expression of MDA5, RIG-I and OAS2 was observed and TLR3 expression was less robust at the protein level upon virus infection (Fig. 5e).

siRNA-mediated knockdown of RNA sensors partially inhibited the cytokine and chemokine responses of human mast cells to VSV infection and affected their replication

To investigate whether RNA sensors are responsible for the recognition and activation of human mast cells during viral infection, we used specific siRNAs to knockdown the respective RNA sensors and examined the anti-viral cytokine and chemokine responses. The specific knockdown of each RNA sensor by the corresponding siRNA was confirmed 24 h after siRNA transfection (data not shown). The cells were stimulated by VSV infection, and their cytokine and chemokine expression was measured by real-time PCR. Although we observed significant but minimal inhibition of cytokine-chemokine induction by the knockdown of individual receptors, the simultaneous knockdown of MDA5, RIG-I and TLR3, along with the knockdown of OAS1–3, significantly inhibited the cytokine and chemokine responses to VSV infection (Fig. 6a). Additionally, we examined the titer of VSV by counting plaque forming units (PFUs) using Vero cells infected with the supernatant of VSV-infected LAD2 cells. Consistent with the results of the cytokine and chemokine analysis, the knockdown of MDA5, RIG-I and TLR3 and OAS1–3 significantly increased VSV replication, suggesting that mast cells exert direct anti-viral responses via the recognition of viral products using these receptors (Fig. 6b). Further investigation suggested that all of these receptors, except OAS1, were responsible for the anti-viral responses of human mast cells infected with VSV (Fig. 6c).

Discussion

We previously demonstrated that murine bone marrow-derived mast cells expressed several viral recognition receptors and responded to viruses via these receptors by producing anti-viral cytokines and chemokines [19]. Mast cells are tissue resident cells derived from hematopoietic stem cells in the bone marrow, which enter the peripheral blood as immature cells and differentiate into mature mast cells under the influence of the surrounding microenvironment [1]. Mast cells display various phenotypes; in particular, these cells differentially express protease in their granules according to the host source and to the associated tissue or organ. Mast cells also possess a complement of PRRs that differentially respond against pathogen-associated molecular patterns (PAMPs) [2022]. Several reports have defined the differential usage of PRRs between human and murine mast cells for the recognition of PAMPs. Thus, in this study, we aimed to identify the virus recognition receptors and their roles in human mast cell activation during virus infection.
We performed PCR array analysis of human mast cells upon VSV infection to identify the genes that are regulated during this infection. As expected, we observed the up-regulation of several genes encoding anti-viral cytokine, chemokines and receptors involved in the recognition of viruses (Table 1). The low expression levels of TLR3 compared with other RLRs were similar in murine mast cells; however, unlike in murine mast cells [19, 23], the lack of TLR3 in human mast cells slightly but significantly reduced the cytokine and chemokine responses and affected VSV replication (Fig. 6), suggesting that TLR3 is also responsible for the recognition of viral products during VSV infection in humans. Although TLR3 has repeatedly been assumed to play an important role in the activation of human and rodent mast cells by poly I:C and several viruses [9, 24], this phenomenon varied among the populations of examined mast cells [25]. Because TLR3-mediated mast cell activation is required for the recruitment of CD8+ T cells to the site of viral infection in some situations [15, 26], TLR3-mediated systems provide different contributions depending on the type of virus and on the route of infection. Although we could detect increased TLR3 expression in VSV-stimulated human mast cells, no significant differences in the expression of other TLRs were observed (data not shown).
RLRs are broadly expressed in most tissues, and their expression is typically maintained at low levels in resting cells but greatly increased after IFN exposure and viral infection [27, 28]. We observed a drastic increase in the RNA sensors RIG-I and MDA5 in VSV-stimulated human mast cells. A similar up-regulation of the mRNA and protein levels of RLRs, including multiple interferon-stimulated genes, was also observed in human mast cells in response to antibody-enhanced Dengue virus or Sendai virus infection [25, 29]. VSV, which is a member of the Rhabdovirus family, contains a single-stranded negative-sense RNA genome. The mechanism by which the innate immune system detects VSV has been thoroughly investigated [3034]; RIG-I mediates a specific response to VSV, as well as to many other negative-strand viruses, such as influenza and Sendai virus, and some positive-strand RNA viruses, such as Japanese encephalitis virus [35]. However, the previous results of our group or others using mouse mast cells have suggested that the responses against VSV were not solely mediated by RIG-I [19, 36, 37]. Our present results suggest that human mast cells responded to VSV via MDA5 and TLR3, in addition to RIG-I (Fig. 6a). Thus, the dependency on different RLRs to recognize different viruses appears to be variable among cells, and the various receptors may provide different contributions depending on the route of infection.
The present study demonstrated for the first time that human mast cells express and up-regulate OAS upon VSV infection (Table 1; Fig. 5). OAS proteins are a family of anti-viral restriction factors that target a wide range of RNA and DNA viruses [38]. These proteins function as intracellular dsRNA sensors that undergo a conformational change upon binding to dsRNA and that are activated to synthesize 2’-5’-linked oligoadenylates (2-5As). Then, 2-5As activate the latent form of RNase L, which leads to viral RNA degradation [39]. In recent years, the contributions of RNase L and OAS to innate immunity have become increasingly apparent [40]. Because the small RNA cleavage products produced by RNase L during viral infections can reportedly signal to the RIG-I receptor [41], our finding that the anti-viral effect mediated by OAS-RNase L was either dependent on or independent of the RIG-receptor system in our studies is notable. siRNA knockdown experiments indicated that OAS knockdown significantly reduced cytokine and chemokine production and virus replication; however, the effect was less complete than the knockdown effects of cells treated with siRNAs for RLRs and TLR3, suggesting that the RIG-I receptor could recognize also the degraded RNA independent of the OAS system (Fig. 6).
The identified OAS genes in humans have been named OAS1, OAS2, OAS3 and OASL (OAS-like) [42]. OAS1–3 have considerable homology to each other, with OAS1, OAS2 and OAS3 encoding one, two and three “OAS” domains, respectively [40]. However, the roles of OAS1–3 in individual viral infections have not been fully investigated [42]. In the PCR array, although we primarily focused on OAS2, we also investigated the expression of OAS 1 and 3. Because the relative expression of OAS2 in un-stimulated human mast cells was low compared with that of OAS1 and OAS3, the drastic increase in OAS2 expression upon VSV infection was obvious. The knockdown experiments for each OAS revealed that OAS2 and 3, but not OAS1, were responsible for the anti-viral effects of human mast cells.
Most pathogens appear to induce lipid mediator and cytokine secretion but not degranulation [7]. We found that VSV infection did not induce β-hexosaminidase release in human mast cells, which is also consistent with previous reports [13, 43] but differs from the results obtained with mouse mast cells [19], in which we observed the degranulation of mouse mast cells after 6 h, but not after 1 h, of VSV infection in the absence of MDA5, RIG-I, PKR and TLR3 [19]. Further studies are required to determine the mechanisms of virus recognition that lead to mast cell degranulation, as well as the differences between human and mouse mast cells.
The induction of type 1 IFNs, including IFN-α and IFN-β, by viruses and other pathogens is of crucial importance for innate immunity [44]. In addition, we observed the up-regulation of several chemokines in human mast cells upon VSV infection (Table 1, Figs. 2, 3). CXCL10, which is a chemokine associated with type 1 T cell responses, is highly expressed in virus-infected cells and is involved in regulating the migration of effector T cells at the sites of inflammation through binding to the CXCR3 receptor [45]. Although the actual role of CXCL11 in viral infections remains unclear, the peak level of CXCL11 mRNA coincides with the peak of viremia and the CXCL11 protein was reported to inhibit viral growth [46]. CXCL11 also directly inhibits respiratory adenovirus serotypes Ad3 and Ad5 [47]. CCL5 is a chemokine and an important inflammatory mediator; CCL5 regulates diseases due to viral infection by signaling via CCR5 on CD8(+) T cells and modulates anti-viral activities [48]. Although we could not detect protein production, a significant increase in IL-15 mRNA was observed upon VSV infection. IL-15 is a well-known cytokine crucial for the development and function of NK cells, which are described as guardians for the detection and clearance of virus-infected cells [49].
Thus, the present findings strengthen the role of mast cells as key responders of the immune response during the early stages of viral infection via their ability to directly recognize and quickly respond to a virus by the rapid production of anti-viral cytokines and chemokines using RLRs and OAS-RNase L, in addition to TLR3. Notably, both allergens and microbial antigens can trigger mast cell activation, and allergic or autoimmune diseases, where mast cells play important roles in their pathogenesis, are often exacerbated by viruses such as rhinovirus [50, 51]. Thus, this knowledge could potentially be exploited therapeutically to modulate host antiviral immunity by targeting these pathways in mast cells.

Acknowledgments

This work was supported in part by the MEXT-Supported Program for the Strategic Research Foundation at Private Universities (Atopy (Allergy) Research Center, Juntendo University). The authors thank Drs. K. Honda and H. Yanai for providing viruses. We also thank the members of the Atopy Research Center and the Department of Immunology for fruitful discussions and technical assistance and Ms. Michiyo Matsumoto for secretarial assistance.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Voehringer D. Protective and pathological roles of mast cells and basophils. Nat Rev Immunol. 2013;13:362–75.CrossRefPubMed Voehringer D. Protective and pathological roles of mast cells and basophils. Nat Rev Immunol. 2013;13:362–75.CrossRefPubMed
2.
Zurück zum Zitat Galli SJ, Tsai M. Mast cells: versatile regulators of inflammation, tissue remodeling, host defense and homeostasis. J Dermatol Sci. 2008;49:7–19.CrossRefPubMedCentralPubMed Galli SJ, Tsai M. Mast cells: versatile regulators of inflammation, tissue remodeling, host defense and homeostasis. J Dermatol Sci. 2008;49:7–19.CrossRefPubMedCentralPubMed
4.
Zurück zum Zitat Abraham SN, St John AL. Mast cell-orchestrated immunity to pathogens. Nat Rev Immunol. 2010;10:440–52.CrossRefPubMed Abraham SN, St John AL. Mast cell-orchestrated immunity to pathogens. Nat Rev Immunol. 2010;10:440–52.CrossRefPubMed
5.
Zurück zum Zitat Supajatura V, Ushio H, Nakao A, Akira S, Okumura K, Ra C, Ogawa H. Differential responses of mast cell Toll-like receptors 2 and 4 in allergy and innate immunity. J Clin Invest. 2002;109:1351–9.CrossRefPubMedCentralPubMed Supajatura V, Ushio H, Nakao A, Akira S, Okumura K, Ra C, Ogawa H. Differential responses of mast cell Toll-like receptors 2 and 4 in allergy and innate immunity. J Clin Invest. 2002;109:1351–9.CrossRefPubMedCentralPubMed
6.
Zurück zum Zitat Supajatura V, Ushio H, Nakao A, Okumura K, Ra C, Ogawa H. Protective roles of mast cells against enterobacterial infection are mediated by Toll-like receptor 4. J Immunol. 2001;167:2250–6.CrossRefPubMed Supajatura V, Ushio H, Nakao A, Okumura K, Ra C, Ogawa H. Protective roles of mast cells against enterobacterial infection are mediated by Toll-like receptor 4. J Immunol. 2001;167:2250–6.CrossRefPubMed
8.
Zurück zum Zitat Yoneyama M, Fujita T. Recognition of viral nucleic acids in innate immunity. Rev Med Virol. 2010;20:4–22.CrossRefPubMed Yoneyama M, Fujita T. Recognition of viral nucleic acids in innate immunity. Rev Med Virol. 2010;20:4–22.CrossRefPubMed
9.
Zurück zum Zitat Kulka M, Alexopoulou L, Flavell RA, Metcalfe DD. Activation of mast cells by double-stranded RNA: evidence for activation through Toll-like receptor 3. J Allergy Clin Immunol. 2004;114:174–82.CrossRefPubMed Kulka M, Alexopoulou L, Flavell RA, Metcalfe DD. Activation of mast cells by double-stranded RNA: evidence for activation through Toll-like receptor 3. J Allergy Clin Immunol. 2004;114:174–82.CrossRefPubMed
10.
Zurück zum Zitat Dietrich N, Rohde M, Geffers R, Kroger A, Hauser H, Weiss S, Gekara NO. Mast cells elicit proinflammatory but not type I interferon responses upon activation of TLRs by bacteria. Proc Natl Acad Sci USA. 2010;107:8748–53.CrossRefPubMedCentralPubMed Dietrich N, Rohde M, Geffers R, Kroger A, Hauser H, Weiss S, Gekara NO. Mast cells elicit proinflammatory but not type I interferon responses upon activation of TLRs by bacteria. Proc Natl Acad Sci USA. 2010;107:8748–53.CrossRefPubMedCentralPubMed
11.
Zurück zum Zitat Marshall JS, King CA, McCurdy JD. Mast cell cytokine and chemokine responses to bacterial and viral infection. Curr Pharm Des. 2003;9:11–24.CrossRefPubMed Marshall JS, King CA, McCurdy JD. Mast cell cytokine and chemokine responses to bacterial and viral infection. Curr Pharm Des. 2003;9:11–24.CrossRefPubMed
12.
Zurück zum Zitat Patella V, Florio G, Petraroli A, Marone G. HIV-1 gp120 induces IL-4 and IL-13 release from human Fc epsilon RI+ cells through interaction with the VH3 region of IgE. J Immunol. 2000;164:589–95.CrossRefPubMed Patella V, Florio G, Petraroli A, Marone G. HIV-1 gp120 induces IL-4 and IL-13 release from human Fc epsilon RI+ cells through interaction with the VH3 region of IgE. J Immunol. 2000;164:589–95.CrossRefPubMed
14.
Zurück zum Zitat Sugiyama K. Histamine release from rat mast cells induced by Sendai virus. Nature. 1977;270:614–5.CrossRefPubMed Sugiyama K. Histamine release from rat mast cells induced by Sendai virus. Nature. 1977;270:614–5.CrossRefPubMed
15.
Zurück zum Zitat Orinska Z, Bulanova E, Budagian V, Metz M, Maurer M, Bulfone-Paus S. TLR3-induced activation of mast cells modulates CD8+ T-cell recruitment. Blood. 2005;106:978–87.CrossRefPubMed Orinska Z, Bulanova E, Budagian V, Metz M, Maurer M, Bulfone-Paus S. TLR3-induced activation of mast cells modulates CD8+ T-cell recruitment. Blood. 2005;106:978–87.CrossRefPubMed
16.
Zurück zum Zitat Burke SM, Issekutz TB, Mohan K, Lee PW, Shmulevitz M, Marshall JS. Human mast cell activation with virus-associated stimuli leads to the selective chemotaxis of natural killer cells by a CXCL8-dependent mechanism. Blood. 2008;111:5467–76.CrossRefPubMed Burke SM, Issekutz TB, Mohan K, Lee PW, Shmulevitz M, Marshall JS. Human mast cell activation with virus-associated stimuli leads to the selective chemotaxis of natural killer cells by a CXCL8-dependent mechanism. Blood. 2008;111:5467–76.CrossRefPubMed
17.
Zurück zum Zitat Stelekati E, Bahri R, D’Orlando O, Orinska Z, Mittrucker HW, Langenhaun R, Glatzel M, Bollinger A, Paus R, Bulfone-Paus S. Mast cell-mediated antigen presentation regulates CD8+ T cell effector functions. Immunity. 2009;31:665–76.CrossRefPubMed Stelekati E, Bahri R, D’Orlando O, Orinska Z, Mittrucker HW, Langenhaun R, Glatzel M, Bollinger A, Paus R, Bulfone-Paus S. Mast cell-mediated antigen presentation regulates CD8+ T cell effector functions. Immunity. 2009;31:665–76.CrossRefPubMed
18.
Zurück zum Zitat Aoki R, Kawamura T, Goshima F, Ogawa Y, Nakae S, Nakao A, Moriishi K, Nishiyama Y, Shimada S. Mast cells play a key role in host defense against herpes simplex virus infection through TNF-alpha and IL-6 production. J Invest Dermatol. 2013;133:2170–9.CrossRefPubMed Aoki R, Kawamura T, Goshima F, Ogawa Y, Nakae S, Nakao A, Moriishi K, Nishiyama Y, Shimada S. Mast cells play a key role in host defense against herpes simplex virus infection through TNF-alpha and IL-6 production. J Invest Dermatol. 2013;133:2170–9.CrossRefPubMed
19.
Zurück zum Zitat Fukuda M, Ushio H, Kawasaki J, Niyonsaba F, Takeuchi M, Baba T, Hiramatsu K, Okumura K, Ogawa H. Expression and functional characterization of retinoic acid-inducible gene-I-like receptors of mast cells in response to viral infection. J Innate Immun. 2013;5:163–73.CrossRefPubMed Fukuda M, Ushio H, Kawasaki J, Niyonsaba F, Takeuchi M, Baba T, Hiramatsu K, Okumura K, Ogawa H. Expression and functional characterization of retinoic acid-inducible gene-I-like receptors of mast cells in response to viral infection. J Innate Immun. 2013;5:163–73.CrossRefPubMed
20.
Zurück zum Zitat Bonini S, Micera A, Iovieno A, Lambiase A. Expression of Toll-like receptors in healthy and allergic conjunctiva. Ophthalmology. 2005;112:1528; discussion 48–49.CrossRefPubMed Bonini S, Micera A, Iovieno A, Lambiase A. Expression of Toll-like receptors in healthy and allergic conjunctiva. Ophthalmology. 2005;112:1528; discussion 48–49.CrossRefPubMed
21.
Zurück zum Zitat McCurdy JD, Olynych TJ, Maher LH, Marshall JS. Cutting edge: distinct Toll-like receptor 2 activators selectively induce different classes of mediator production from human mast cells. J Immunol. 2003;170:1625–9.CrossRefPubMed McCurdy JD, Olynych TJ, Maher LH, Marshall JS. Cutting edge: distinct Toll-like receptor 2 activators selectively induce different classes of mediator production from human mast cells. J Immunol. 2003;170:1625–9.CrossRefPubMed
22.
Zurück zum Zitat Varadaradjalou S, Feger F, Thieblemont N, Hamouda NB, Pleau JM, Dy M, Arock M. Toll-like receptor 2 (TLR2) and TLR4 differentially activate human mast cells. Eur J Immunol. 2003;33:899–906.CrossRefPubMed Varadaradjalou S, Feger F, Thieblemont N, Hamouda NB, Pleau JM, Dy M, Arock M. Toll-like receptor 2 (TLR2) and TLR4 differentially activate human mast cells. Eur J Immunol. 2003;33:899–906.CrossRefPubMed
23.
Zurück zum Zitat Edelmann KH, Richardson-Burns S, Alexopoulou L, Tyler KL, Flavell RA, Oldstone MB. Does Toll-like receptor 3 play a biological role in virus infections? Virology. 2004;322:231–8.CrossRefPubMed Edelmann KH, Richardson-Burns S, Alexopoulou L, Tyler KL, Flavell RA, Oldstone MB. Does Toll-like receptor 3 play a biological role in virus infections? Virology. 2004;322:231–8.CrossRefPubMed
24.
Zurück zum Zitat Matsushima H, Yamada N, Matsue H, Shimada S. TLR3-, TLR7-, and TLR9-mediated production of proinflammatory cytokines and chemokines from murine connective tissue type skin-derived mast cells but not from bone marrow-derived mast cells. J Immunol. 2004;173:531–41.CrossRefPubMed Matsushima H, Yamada N, Matsue H, Shimada S. TLR3-, TLR7-, and TLR9-mediated production of proinflammatory cytokines and chemokines from murine connective tissue type skin-derived mast cells but not from bone marrow-derived mast cells. J Immunol. 2004;173:531–41.CrossRefPubMed
25.
Zurück zum Zitat Brown MG, McAlpine SM, Huang YY, Haidl ID, Al-Afif A, Marshall JS, Anderson R. RNA sensors enable human mast cell anti-viral chemokine production and IFN-mediated protection in response to antibody-enhanced dengue virus infection. PLoS ONE. 2012;7:e34055.CrossRefPubMedCentralPubMed Brown MG, McAlpine SM, Huang YY, Haidl ID, Al-Afif A, Marshall JS, Anderson R. RNA sensors enable human mast cell anti-viral chemokine production and IFN-mediated protection in response to antibody-enhanced dengue virus infection. PLoS ONE. 2012;7:e34055.CrossRefPubMedCentralPubMed
26.
Zurück zum Zitat Schulz O, Diebold SS, Chen M, Naslund TI, Nolte MA, Alexopoulou L, Azuma YT, Flavell RA, Liljestrom P, Reis e Sousa C. Toll-like receptor 3 promotes cross-priming to virus-infected cells. Nature. 2005;433:887–92.CrossRefPubMed Schulz O, Diebold SS, Chen M, Naslund TI, Nolte MA, Alexopoulou L, Azuma YT, Flavell RA, Liljestrom P, Reis e Sousa C. Toll-like receptor 3 promotes cross-priming to virus-infected cells. Nature. 2005;433:887–92.CrossRefPubMed
27.
Zurück zum Zitat Sumpter R Jr, Loo YM, Foy E, Li K, Yoneyama M, Fujita T, Lemon SM, Gale M Jr. Regulating intracellular antiviral defense and permissiveness to hepatitis C virus RNA replication through a cellular RNA helicase, RIG-I. J Virol. 2005;79:2689–99.CrossRefPubMedCentralPubMed Sumpter R Jr, Loo YM, Foy E, Li K, Yoneyama M, Fujita T, Lemon SM, Gale M Jr. Regulating intracellular antiviral defense and permissiveness to hepatitis C virus RNA replication through a cellular RNA helicase, RIG-I. J Virol. 2005;79:2689–99.CrossRefPubMedCentralPubMed
28.
Zurück zum Zitat Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi M, Taira K, Akira S, Fujita T. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol. 2004;5:730–7.CrossRefPubMed Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi M, Taira K, Akira S, Fujita T. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol. 2004;5:730–7.CrossRefPubMed
29.
Zurück zum Zitat Lappalainen J, Rintahaka J, Kovanen PT, Matikainen S, Eklund KK. Intracellular RNA recognition pathway activates strong anti-viral response in human mast cells. Clin Exp Immunol. 2013;172:121–8.CrossRefPubMedCentralPubMed Lappalainen J, Rintahaka J, Kovanen PT, Matikainen S, Eklund KK. Intracellular RNA recognition pathway activates strong anti-viral response in human mast cells. Clin Exp Immunol. 2013;172:121–8.CrossRefPubMedCentralPubMed
30.
Zurück zum Zitat Georgel P, Jiang Z, Kunz S, Janssen E, Mols J, Hoebe K, Bahram S, Oldstone MB, Beutler B. Vesicular stomatitis virus glycoprotein G activates a specific antiviral Toll-like receptor 4-dependent pathway. Virology. 2007;362:304–13.CrossRefPubMed Georgel P, Jiang Z, Kunz S, Janssen E, Mols J, Hoebe K, Bahram S, Oldstone MB, Beutler B. Vesicular stomatitis virus glycoprotein G activates a specific antiviral Toll-like receptor 4-dependent pathway. Virology. 2007;362:304–13.CrossRefPubMed
31.
Zurück zum Zitat Yoneyama M, Kikuchi M, Matsumoto K, Imaizumi T, Miyagishi M, Taira K, Foy E, Loo YM, Gale M Jr, Akira S, Yonehara S, Kato A, Fujita T. Shared and unique functions of the DExD/H-box helicases RIG-I, MDA5, and LGP2 in antiviral innate immunity. J Immunol. 2005;175:2851–8.CrossRefPubMed Yoneyama M, Kikuchi M, Matsumoto K, Imaizumi T, Miyagishi M, Taira K, Foy E, Loo YM, Gale M Jr, Akira S, Yonehara S, Kato A, Fujita T. Shared and unique functions of the DExD/H-box helicases RIG-I, MDA5, and LGP2 in antiviral innate immunity. J Immunol. 2005;175:2851–8.CrossRefPubMed
32.
Zurück zum Zitat Kato H, Sato S, Yoneyama M, Yamamoto M, Uematsu S, Matsui K, Tsujimura T, Takeda K, Fujita T, Takeuchi O, Akira S. Cell type-specific involvement of RIG-I in antiviral response. Immunity. 2005;23:19–28.CrossRefPubMed Kato H, Sato S, Yoneyama M, Yamamoto M, Uematsu S, Matsui K, Tsujimura T, Takeda K, Fujita T, Takeuchi O, Akira S. Cell type-specific involvement of RIG-I in antiviral response. Immunity. 2005;23:19–28.CrossRefPubMed
33.
Zurück zum Zitat Lund JM, Alexopoulou L, Sato A, Karow M, Adams NC, Gale NW, Iwasaki A, Flavell RA. Recognition of single-stranded RNA viruses by Toll-like receptor 7. Proc Natl Acad Sci USA. 2004;101:5598–603.CrossRefPubMedCentralPubMed Lund JM, Alexopoulou L, Sato A, Karow M, Adams NC, Gale NW, Iwasaki A, Flavell RA. Recognition of single-stranded RNA viruses by Toll-like receptor 7. Proc Natl Acad Sci USA. 2004;101:5598–603.CrossRefPubMedCentralPubMed
34.
Zurück zum Zitat Pichlmair A, Schulz O, Tan CP, Naslund TI, Liljestrom P, Weber F, Reis e Sousa C. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5’-phosphates. Science. 2006;314:997–1001.CrossRefPubMed Pichlmair A, Schulz O, Tan CP, Naslund TI, Liljestrom P, Weber F, Reis e Sousa C. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5’-phosphates. Science. 2006;314:997–1001.CrossRefPubMed
35.
Zurück zum Zitat Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, Uematsu S, Jung A, Kawai T, Ishii KJ, Yamaguchi O, Otsu K, Tsujimura T, Koh CS, Reis e Sousa C, Matsuura Y, Fujita T, Akira S. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006;441:101–5.CrossRefPubMed Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, Uematsu S, Jung A, Kawai T, Ishii KJ, Yamaguchi O, Otsu K, Tsujimura T, Koh CS, Reis e Sousa C, Matsuura Y, Fujita T, Akira S. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006;441:101–5.CrossRefPubMed
36.
Zurück zum Zitat St John AL, Rathore AP, Yap H, Ng ML, Metcalfe DD, Vasudevan SG, Abraham SN. Immune surveillance by mast cells during dengue infection promotes natural killer (NK) and NKT-cell recruitment and viral clearance. Proc Natl Acad Sci USA. 2011;108:9190–5.CrossRefPubMedCentralPubMed St John AL, Rathore AP, Yap H, Ng ML, Metcalfe DD, Vasudevan SG, Abraham SN. Immune surveillance by mast cells during dengue infection promotes natural killer (NK) and NKT-cell recruitment and viral clearance. Proc Natl Acad Sci USA. 2011;108:9190–5.CrossRefPubMedCentralPubMed
37.
Zurück zum Zitat Saito T, Gale M Jr. Principles of intracellular viral recognition. Curr Opin Immunol. 2007;19:17–23.CrossRefPubMed Saito T, Gale M Jr. Principles of intracellular viral recognition. Curr Opin Immunol. 2007;19:17–23.CrossRefPubMed
38.
Zurück zum Zitat Kristiansen H, Gad HH, Eskildsen-Larsen S, Despres P, Hartmann R. The oligoadenylate synthetase family: an ancient protein family with multiple antiviral activities. J Interferon Cytokine Res. 2011;31:41–7.CrossRefPubMed Kristiansen H, Gad HH, Eskildsen-Larsen S, Despres P, Hartmann R. The oligoadenylate synthetase family: an ancient protein family with multiple antiviral activities. J Interferon Cytokine Res. 2011;31:41–7.CrossRefPubMed
39.
41.
42.
Zurück zum Zitat Hovanessian AG, Justesen J. The human 2′-5′ oligoadenylate synthetase family: unique interferon-inducible enzymes catalyzing 2′-5′ instead of 3′-5′ phosphodiester bond formation. Biochimie. 2007;89:779–88.CrossRefPubMed Hovanessian AG, Justesen J. The human 2′-5′ oligoadenylate synthetase family: unique interferon-inducible enzymes catalyzing 2′-5′ instead of 3′-5′ phosphodiester bond formation. Biochimie. 2007;89:779–88.CrossRefPubMed
43.
Zurück zum Zitat Kulka M, Metcalfe DD. Isolation of tissue mast cells. Curr Protoc Immunol. 2010;Chapter 7:Unit 7.25. Kulka M, Metcalfe DD. Isolation of tissue mast cells. Curr Protoc Immunol. 2010;Chapter 7:Unit 7.25.
44.
Zurück zum Zitat Takeuchi O, Akira S. Recognition of viruses by innate immunity. Immunol Rev. 2007;220:214–24.CrossRefPubMed Takeuchi O, Akira S. Recognition of viruses by innate immunity. Immunol Rev. 2007;220:214–24.CrossRefPubMed
45.
Zurück zum Zitat Christensen JE, de Lemos C, Moos T, Christensen JP, Thomsen AR. CXCL10 is the key ligand for CXCR3 on CD8+ effector T cells involved in immune surveillance of the lymphocytic choriomeningitis virus-infected central nervous system. J Immunol. 2006;176:4235–43.CrossRefPubMed Christensen JE, de Lemos C, Moos T, Christensen JP, Thomsen AR. CXCL10 is the key ligand for CXCR3 on CD8+ effector T cells involved in immune surveillance of the lymphocytic choriomeningitis virus-infected central nervous system. J Immunol. 2006;176:4235–43.CrossRefPubMed
46.
Zurück zum Zitat Hamilton NH, Mahalingam S, Banyer JL, Ramshaw IA, Thomson SA. A recombinant vaccinia virus encoding the interferon-inducible T-cell alpha chemoattractant is attenuated in vivo. Scand J Immunol. 2004;59:246–54.CrossRefPubMed Hamilton NH, Mahalingam S, Banyer JL, Ramshaw IA, Thomson SA. A recombinant vaccinia virus encoding the interferon-inducible T-cell alpha chemoattractant is attenuated in vivo. Scand J Immunol. 2004;59:246–54.CrossRefPubMed
47.
Zurück zum Zitat Harvey SA, Romanowski EG, Yates KA, Gordon YJ. Adenovirus-directed ocular innate immunity: the role of conjunctival defensin-like chemokines (IP-10, I-TAC) and phagocytic human defensin-alpha. Invest Ophthalmol Vis Sci. 2005;46:3657–65.CrossRefPubMed Harvey SA, Romanowski EG, Yates KA, Gordon YJ. Adenovirus-directed ocular innate immunity: the role of conjunctival defensin-like chemokines (IP-10, I-TAC) and phagocytic human defensin-alpha. Invest Ophthalmol Vis Sci. 2005;46:3657–65.CrossRefPubMed
48.
Zurück zum Zitat Glass WG, Rosenberg HF, Murphy PM. Chemokine regulation of inflammation during acute viral infection. Curr Opin Allergy Clin Immunol. 2003;3:467–73.CrossRefPubMed Glass WG, Rosenberg HF, Murphy PM. Chemokine regulation of inflammation during acute viral infection. Curr Opin Allergy Clin Immunol. 2003;3:467–73.CrossRefPubMed
50.
Zurück zum Zitat Munz C, Lunemann JD, Getts MT, Miller SD. Antiviral immune responses: triggers of or triggered by autoimmunity? Nat Rev Immunol. 2009;9:246–58.CrossRefPubMedCentralPubMed Munz C, Lunemann JD, Getts MT, Miller SD. Antiviral immune responses: triggers of or triggered by autoimmunity? Nat Rev Immunol. 2009;9:246–58.CrossRefPubMedCentralPubMed
51.
Zurück zum Zitat Kallal LE, Lukacs NW. The role of chemokines in virus-associated asthma exacerbations. Curr Allergy Asthma Rep. 2008;8:443–50.CrossRefPubMed Kallal LE, Lukacs NW. The role of chemokines in virus-associated asthma exacerbations. Curr Allergy Asthma Rep. 2008;8:443–50.CrossRefPubMed
Metadaten
Titel
Roles of retinoic acid-inducible gene-I-like receptors (RLRs), Toll-like receptor (TLR) 3 and 2′-5′ oligoadenylate synthetase as viral recognition receptors on human mast cells in response to viral infection
verfasst von
Mizuho Tsutsui-Takeuchi
Hiroko Ushio
Minoru Fukuda
Takahiko Yamada
François Niyonsaba
Ko Okumura
Hideoki Ogawa
Shigaku Ikeda
Publikationsdatum
01.03.2015
Verlag
Springer US
Erschienen in
Immunologic Research / Ausgabe 3/2015
Print ISSN: 0257-277X
Elektronische ISSN: 1559-0755
DOI
https://doi.org/10.1007/s12026-014-8617-x

Weitere Artikel der Ausgabe 3/2015

Immunologic Research 3/2015 Zur Ausgabe

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Klinik aktuell Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Nur selten Nachblutungen nach Abszesstonsillektomie

03.05.2024 Tonsillektomie Nachrichten

In einer Metaanalyse von 18 Studien war die Rate von Nachblutungen nach einer Abszesstonsillektomie mit weniger als 7% recht niedrig. Nur rund 2% der Behandelten mussten nachoperiert werden. Die Therapie scheint damit recht sicher zu sein.

Rezidivierender Peritonsillarabszess nach Oralsex

02.05.2024 Peritonsillarabszess Kasuistik

Die erotischen Dimensionen von Peritonsillarabszessen scheinen eng begrenzt zu sein. Das heißt aber nicht, solche Abszesse und Erotik hätten nichts miteinander gemein, wie ein Fallbericht verdeutlicht.

Update HNO

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.