Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Clin Exp Immunol ; 215(1): 37-46, 2024 01 09.
Article En | MEDLINE | ID: mdl-37583293

Staphylococcus aureus (SA) and its exotoxins activate eosinophils (Eos) and mast cells (MCs) via CD48, a GPI-anchored receptor belonging to the signaling lymphocytes activation molecules (SLAM) family. 2B4 (CD244), an immuno-regulatory transmembrane receptor also belonging to the SLAM family, is the high-affinity ligand for CD48. 2B4 is expressed on several leukocytes including NK cells, T cells, basophils, monocytes, dendritic cells (DCs), and Eos. In the Eos and MCs crosstalk carried out by physical and soluble interactions (named the 'allergic effector unit', AEU), 2B4-CD48 binding plays a central role. As CD48 and 2B4 share some structural characteristics and SA colonization accompanies most of the allergic diseases, we hypothesized that SA exotoxins (e.g. Staphylococcus enterotoxin B, SEB) can also bind and activate 2B4 and thereby possibly further aggravate inflammation. To check our hypothesis, we used in vitro, in silico, and in vivo methods. By enzyme-linked immunosorbent assay (ELISA), flow cytometry (FC), fluorescence microscopy, and microscale thermophoresis, we have shown that SEB can bind specifically to 2B4. By Eos short- and long-term activation assays, we confirmed the functionality of the SEB-2B4 interaction. Using computational modeling, we identified possible SEB-binding sites on human and mouse 2B4. Finally, in vivo, in an SEB-induced peritonitis model, 2B4-KO mice showed a significant reduction of inflammatory features compared with WT mice. Altogether, the results of this study confirm that 2B4 is an important receptor in SEB-mediated inflammation, and therefore a role is suggested for 2B4 in SA associated inflammatory conditions.


Hypersensitivity , Staphylococcus aureus , Animals , Humans , Mice , CD48 Antigen/metabolism , Exotoxins , Inflammation , Signaling Lymphocytic Activation Molecule Family , Staphylococcus aureus/metabolism
3.
Ann Allergy Asthma Immunol ; 130(2): 245-253.e9, 2023 02.
Article En | MEDLINE | ID: mdl-36280100

BACKGROUND: Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can progress into a severe form of acute lung injury. The cosignaling receptor cluster of differentiation 48 (CD48) exists in membrane-bound (mCD48) and soluble (sCD48) forms and has been reported to be implicated in antiviral immunity and dysregulated in several inflammatory conditions. Therefore, CD48 dysregulation may be a putative feature in COVID-19-associated inflammation that deserves consideration. OBJECTIVE: To analyze CD48 expression in lung autopsies and peripheral blood leukocytes and sera of patients with COVID-19. The expression of the CD48 ligand 2B4 on the membrane of peripheral blood leukocytes was also assessed. METHODS: Twenty-eight lung tissue samples obtained from COVID-19 autopsies were assessed for CD48 expression using gene expression profiling immunohistochemistry (HTG autoimmune panel). Peripheral whole blood was collected from 111 patients with COVID-19, and the expression of mCD48 and of membrane-bound 2B4 was analyzed by flow cytometry. Serum levels of sCD48 were assessed by enzyme-linked immunosorbent assay. RESULTS: Lung tissue of patients with COVID-19 showed increased CD48 messenger RNA expression and infiltration of CD48+ lymphocytes. In the peripheral blood, mCD48 was considerably increased on all evaluated cell types. In addition, sCD48 levels were significantly higher in patients with COVID-19, independently of disease severity. CONCLUSION: Considering the changes of mCD48 and sCD48, a role for CD48 in COVID-19 can be assumed and needs to be further investigated.


COVID-19 , Receptors, Immunologic , Humans , CD48 Antigen/metabolism , SARS-CoV-2 , Inflammation
4.
Cancer Gene Ther ; 29(11): 1676-1685, 2022 11.
Article En | MEDLINE | ID: mdl-35681020

Inhibitory receptors (IRs), such as the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), are cell surface molecules expressed on both normal epithelial, endothelial, and hematopoietic cells and on neoplastic cells. IRs are usually used by cancer cells to inhibit immune cell functions. Thus, CEACAM1 positive tumor cells can interact homophilically with CEACAM1 expressed on T and NK cells to inhibit their antibody-dependent cell-mediated cytotoxicity (ADCC). In this study, we investigated the effect of agonistic/activating anti-CEACAM1 monoclonal antibody (mAb) on melanoma cell lines in vitro and in vivo, following our hypothesis that activation of CEACAM1 on melanoma cells by distinct mAbs may induce inhibition of cancer cell proliferation and/or their death. To address this, we established an activating anti-CEACAM1 mAb (CCM5.01) and characterized its binding to the CEACAM1 receptor. Using this mAb, we assessed the expression of CEACAM1 on four different human melanoma cell lines by western blot and flow cytometry and determined its effect on cell viability in vitro by MTT assay. Furthermore, we evaluated the mAb mechanism of action and found that binding of CEACAM1 with CCM5.01 induced SHP1 phosphorylation and p53 activation resulting in melanoma cell apoptosis. For in vivo studies, a xenograft model of melanoma was performed by injection of Mel-14 cells subcutaneously (s.c.) in SCID/Beige mice followed by intraperitoneal (i.p.) injection of CCM5.01 or of IgG1 isotype control every other day. CCM5.01 treated mice showed a slight but not significant decrease in tumor weight in comparison to the control group. Based on the obtained data, we suggest that activating CEACAM1 on melanoma cells might be a promising novel approach to fight cancers expressing this IR.


Antibodies, Monoclonal , Melanoma , Humans , Mice , Animals , Antibodies, Monoclonal/pharmacology , Cell Adhesion Molecule-1 , Mice, SCID , Cell Line, Tumor , Melanoma/metabolism , Carcinoembryonic Antigen/metabolism
5.
Clin Exp Immunol ; 209(1): 72-82, 2022 07 22.
Article En | MEDLINE | ID: mdl-35467728

Eosinophils are innate immune cells typically associated with allergic and parasitic diseases. However, in recent years, eosinophils have also been ascribed a role in keeping homeostasis and in fighting several infectious diseases. Indeed, these cells circulate as mature cells in the blood and can be quickly recruited to the infected tissue. Moreover, eosinophils have all the necessary cellular equipment such as pattern recognition receptors (PRRs), pro-inflammatory cytokines, anti-bacterial proteins, and DNA traps to fight pathogens and promote an efficient immune response. This review summarizes some of the updated information on the role of eosinophils' direct and indirect mediated interactions with pathogens.


Bacterial Infections , Eosinophils , Mycoses , Virus Diseases , Bacterial Infections/immunology , Cytokines/metabolism , Eosinophils/immunology , Humans , Immunity, Innate , Mycoses/immunology , Receptors, Pattern Recognition/metabolism , Virus Diseases/immunology
6.
Obesity (Silver Spring) ; 29(8): 1272-1278, 2021 08.
Article En | MEDLINE | ID: mdl-34314110

OBJECTIVE: Obesity is a major risk factor that increases morbidity and mortality upon infection. Although type I and type III interferon (IFN)-induced innate immune responses represent the first line of defense against viral infections, their functionality in the context of metabolic disorders remains largely obscure. This study aimed to investigate IFN responses upon respiratory viral infection in obese mice. METHODS: The activation of IFNs as well as IFN regulatory factors (IRFs) upon H3N2 influenza infection in mice upon high-fat-diet feeding was investigated. RESULTS: Influenza infection of obese mice was characterized by higher mortalities. In-depth analysis revealed impaired induction of both type I and type III IFNs as well as markedly reduced IFN responses. Notably, it was found that IRF7 gene expression in obese animals was reduced in homeostasis, and its induction by the virus was strongly attenuated. CONCLUSIONS: The results suggest that the attenuated IRF7 expression and induction are responsible for the reduced expression levels of type I and III IFNs and, thus, for the higher susceptibility and severity of respiratory infections in obese mice.


Influenza A Virus, H3N2 Subtype , Influenza, Human , Animals , Humans , Immunity, Innate , Interferons , Mice , Mice, Obese
8.
Viruses ; 13(5)2021 04 21.
Article En | MEDLINE | ID: mdl-33919410

To establish a productive infection in host cells, viruses often use one or multiple host membrane glycoproteins as their receptors. For Influenza A virus (IAV) such a glycoprotein receptor has not been described, to date. Here we show that IAV is using the host membrane glycoprotein CD66c as a receptor for entry into human epithelial lung cells. Neuraminidase (NA), a viral spike protein, binds to CD66c on the cell surface during IAV entry into the host cells. Lung cells overexpressing CD66c showed an increase in virus binding and subsequent entry into the cell. Upon comparison, CD66c demonstrated higher binding capacity than other membrane glycoproteins (EGFR and DC-SIGN) reported earlier to facilitate IAV entry into host cells. siRNA mediated knockdown of CD66c from lung cells inhibited virus binding on cell surface and entry into cells. Blocking CD66c by antibody on the cell surface resulted in decreased virus entry. We found that CD66c is a specific glycoprotein receptor for influenza A virus that did not affect entry of non-IAV RNA virus (Hepatitis C virus). Finally, IAV pre-incubated with recombinant CD66c protein when administered intranasally in mice showed decreased cytopathic effects in mice lungs. This publication is the first to report CD66c (Carcinoembryonic cell adhesion molecule 6 or CEACAM6) as a glycoprotein receptor for Influenza A virus.


Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Host-Pathogen Interactions , Influenza A virus/physiology , Influenza, Human/metabolism , Influenza, Human/virology , Receptors, Virus/metabolism , Antigens, CD/genetics , Cell Adhesion Molecules/genetics , Cell Line , Flow Cytometry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression , Gene Knockdown Techniques , Genes, Reporter , Humans , Influenza, Human/immunology , Influenza, Human/pathology , RNA, Small Interfering/genetics , Virus Attachment , Virus Internalization , Virus Replication
9.
J Cell Biochem ; 120(4): 6449-6458, 2019 04.
Article En | MEDLINE | ID: mdl-30335904

Neuraminidase protein (NA) of influenza A virus (IAV) is popularly known for its sialidase function to assist in the release of progeny virus. However, involvement of NA in other stages of the IAV life cycle also indicates its multifunctional nature and necessity to interact with other host proteins. Here, we report a host protein-heat shock protein 90 (Hsp90), as a novel interacting partner of IAV NA. A classical yeast two-hybrid screen was conducted to identify a new host interacting partner for NA and the interaction was further validated by coimmunoprecipitation from cells, transiently expressing both proteins and also from IAV-infected cells. Confocal imaging showed that both proteins colocalized in the cytoplasm in transfected host cells. Interestingly, increased levels of NA in the presence of Hsp90 was observed, which tends to decrease if adenosine triphosphatase activity of Hsp90 is inhibited using 17-N-allylamino-17-demethoxygeldanamycin (17AAG). This establishes viral NA as a client protein of host chaperone Hsp90 contributing toward NA's stability via the NA-Hsp90 interaction. This is the first report showing the interaction of NA with Hsp90 and its role in stabilizing viral NA thus preventing it from degradation. Enhanced cell survival in the presence of this interaction was also observed, thus suggesting the requirement of stable viral NA, post-IAV infection, for efficient virus production in infected mammalian cells.


HSP90 Heat-Shock Proteins/metabolism , Influenza A virus/physiology , Influenza, Human/virology , Neuraminidase/chemistry , Neuraminidase/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication , A549 Cells , Cell Survival , Host-Pathogen Interactions , Humans , Influenza, Human/metabolism , Influenza, Human/pathology , Protein Stability
10.
Metabolism ; 69: 171-176, 2017 04.
Article En | MEDLINE | ID: mdl-28285647

AIM: Obesity is accompanied with systemic inflammation and pre-conditions to severe alterations in liver environment and functions. So far, it remains elusive to which extent obesity modulates immune responses during hepatotropic virus infections as well as in autoimmune hepatitis. In this study we investigated the influence of obesity on the intrahepatic immune response, in particular on the function of CD8 T cells as the crucial players in clearance of virus infected hepatocytes. METHODS: We established high fat induced obesity in transgenic mouse models with hepatocyte specific expression of a model antigen (Ova). We investigated the immune response upon adoptive transfer of antigen specific T cells and in mice with continuous thymic output of antigen specific T cells, mimicking the situations upon acute infection and autoimmunity, respectively. RESULTS: Irrespective of the metabolic condition, adoptive T cell transfer resulted in a transient hepatitis with no obvious differences concerning the acute T cell response. In the situation of autoimmunity, we observed a transient hepatitis in lean mice, whereas an extended hepatitis with a reduced antigen clearance capacity was found in obese mice. CONCLUSION: Our results demonstrate that obesity affects T cell function and increases the severity of autoimmune hepatitis while it has no impact on the acute T cell response.


CD8-Positive T-Lymphocytes/immunology , Diet , Hepatocytes/immunology , Liver/immunology , Obesity/immunology , Adoptive Transfer , Animals , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/pathology , Diet, High-Fat , Hepatocytes/pathology , Liver/pathology , Liver Function Tests , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Obesity/pathology , Ovalbumin/immunology , Thymus Gland/cytology , Thymus Gland/immunology
11.
J Nat Prod ; 78(3): 374-80, 2015 Mar 27.
Article En | MEDLINE | ID: mdl-25699574

A new orbitide named ribifolin was isolated and characterized from Jatropha ribifolia using mass spectrometry, NMR spectroscopy, quantitative amino acid analysis, molecular dynamics/simulated annealing, and Raman optical activity measurements and calculations. Ribifolin (1) and its linear form (1a) were synthesized by solid-phase peptide synthesis, followed by evaluation of its antiplasmodial and cytotoxicity activities. Compound 1 was moderately effective (IC50 = 42 µM) against the Plasmodium falciparum strain 3D7.


Antimalarials , Jatropha/chemistry , Peptides, Cyclic , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Antimalarials/isolation & purification , Antimalarials/pharmacology , Drug Screening Assays, Antitumor , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Parasitic Sensitivity Tests , Peptides, Cyclic/chemistry , Peptides, Cyclic/isolation & purification , Peptides, Cyclic/pharmacology , Plant Extracts/chemistry , Solid-Phase Synthesis Techniques
12.
FEBS J ; 281(13): 2899-914, 2014 Jul.
Article En | MEDLINE | ID: mdl-24802111

Influenza A virus (IAV), similar to other viruses, exploits the machinery of human host cells for its survival and replication. We identified α-actinin-4, a host cytoskeletal protein, as an interacting partner of IAV nucleoprotein (NP). We confirmed this interaction using co-immunoprecipitation studies, first in a coupled in vitro transcription-translation assay and then in cells either transiently co-expressing the two proteins or infected with whole IAV. Importantly, the NP-actinin-4 interaction was observed in several IAV subtypes, including the 2009 H1N1 pandemic virus. Moreover, immunofluorescence studies revealed that both NP and actinin-4 co-localized largely around the nucleus and also in the cytoplasmic region of virus-infected A549 cells. Silencing of actinin-4 expression resulted in not only a significant decrease in NP, M2 and NS1 viral protein expression, but also a reduction of both NP mRNA and viral RNA levels, as well as viral titers, 24 h post-infection with IAV, suggesting that actinin-4 was critical for viral replication. Furthermore, actinin-4 depletion reduced the amount of NP localized in the nucleus. Treatment of infected cells with wortmannin, a known inhibitor of actinin-4, led to a decrease in NP mRNA levels and also caused the nuclear retention of NP, further strengthening our previous observations. Taken together, the results of the present study indicate that actinin-4, a novel interacting partner of IAV NP, plays a crucial role in viral replication and this interaction may participate in nuclear localization of NP and/or viral ribonucleoproteins.


Actinin/metabolism , Influenza A virus/physiology , RNA-Binding Proteins/physiology , Viral Core Proteins/physiology , Virus Replication , Actinin/genetics , HEK293 Cells , Host-Pathogen Interactions , Humans , Nucleocapsid Proteins , Protein Interaction Mapping , Protein Transport , Transcriptional Activation
13.
Eur J Med Chem ; 59: 48-53, 2013 Jan.
Article En | MEDLINE | ID: mdl-23202850

A series of 36 new phenylsulfanylmethyl[1,4]naphthoquinones (7-42) were synthesized by a three-component reaction that involves lawsone, the appropriate aldehyde and thiols with variable substitution patterns. These reactions involve the in situ generation of o-quinone methides (o-QM) via Knoevenagel condensation and 1,4-nucleophilic addition under conventional heating or microwave irradiation. The new naphthoquinones obtained by this methodology were shown to have moderate to good in vitro antimalarial activity against Plasmodium falciparum (3D7).


Antimalarials/chemical synthesis , Antimalarials/pharmacology , Naphthoquinones/chemical synthesis , Naphthoquinones/pharmacology , Plasmodium falciparum/drug effects , Sulfhydryl Compounds/chemical synthesis , Cells, Cultured , Flow Cytometry , Humans , Indolequinones/chemical synthesis , Indolequinones/chemistry , Indolequinones/pharmacology , Malaria, Falciparum , Molecular Structure , Naphthoquinones/chemistry , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology
14.
J Biol Chem ; 287(18): 15109-17, 2012 Apr 27.
Article En | MEDLINE | ID: mdl-22396546

The influenza virus neuraminidase (NA) protein primarily aids in the release of progeny virions from infected cells. Here, we demonstrate a novel role for NA in enhancing host cell survival by activating the Src/Akt signaling axis via an interaction with carcinoembryonic antigen-related cell adhesion molecule 6/cluster of differentiation 66c (C6). NA/C6 interaction leads to increased tyrosyl phosphorylation of Src, FAK, Akt, GSK3ß, and Bcl-2, which affects cell survival, proliferation, migration, differentiation, and apoptosis. siRNA-mediated suppression of C6 resulted in a down-regulation of activated Src, FAK, and Akt, increased apoptosis, and reduced expression of viral proteins and viral titers in influenza virus-infected human lung adenocarcinoma epithelial and normal human bronchial epithelial cells. These findings indicate that influenza NA not only aids in the release of progeny virions, but also cell survival during viral replication.


Antigens, CD/metabolism , Bronchi/metabolism , Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , Influenza A virus/physiology , Influenza, Human/metabolism , Neuraminidase/metabolism , Respiratory Mucosa/metabolism , Viral Proteins/metabolism , Antigens, CD/genetics , Apoptosis/genetics , Bronchi/pathology , Bronchi/virology , Cell Adhesion Molecules/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Epithelial Cells/pathology , Epithelial Cells/virology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Influenza, Human/pathology , Influenza, Human/virology , Neuraminidase/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Signal Transduction/genetics , Viral Proteins/genetics , Virus Replication/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
15.
Med Sci Monit ; 17(6): RA148-54, 2011 Jun.
Article En | MEDLINE | ID: mdl-21629204

Influenza viruses comprise a major class of human respiratory pathogens, responsible for causing morbidity and mortality worldwide. Influenza A virus, due to its segmented RNA genome, is highly subject to mutation, resulting in rapid formation of variants. During influenza infection, viral proteins interact with host proteins and exploit a variety of cellular pathways for their own benefit. Influenza virus inhibits the synthesis of these cellular proteins and facilitates expression of its own proteins for viral transcription and replication. Infected cell pathways are hijacked by an array of intracellular signaling cascades such as NF-κB signaling, PI3K/Akt pathway, MAPK pathway, PKC/PKR signaling and TLR/RIG-I signaling cascades. This review presents a research update on the subject and discusses the impact of influenza viral infection on these cell signaling pathways.


Cells/metabolism , Cells/virology , Orthomyxoviridae/metabolism , Signal Transduction , Animals , Cells/enzymology , Humans , Models, Biological
...