Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
mBio ; 12(4): e0097421, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34253053

ABSTRACT

In the coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), more severe outcomes are reported in males than in females, including hospitalizations and deaths. Animal models can provide an opportunity to mechanistically interrogate causes of sex differences in the pathogenesis of SARS-CoV-2. Adult male and female golden Syrian hamsters (8 to 10 weeks of age) were inoculated intranasally with 105 50% tissue culture infective dose (TCID50) of SARS-CoV-2/USA-WA1/2020 and euthanized at several time points during the acute (i.e., virus actively replicating) and recovery (i.e., after the infectious virus has been cleared) phases of infection. There was no mortality, but infected male hamsters experienced greater morbidity, losing a greater percentage of body mass, developed more extensive pneumonia as noted on chest computed tomography, and recovered more slowly than females. Treatment of male hamsters with estradiol did not alter pulmonary damage. Virus titers in respiratory tissues, including nasal turbinates, trachea, and lungs, and pulmonary cytokine concentrations, including interferon-ß (IFN-ß) and tumor necrosis factor-α (TNF-α), were comparable between the sexes. However, during the recovery phase of infection, females mounted 2-fold greater IgM, IgG, and IgA responses against the receptor-binding domain of the spike protein (S-RBD) in both plasma and respiratory tissues. Female hamsters also had significantly greater IgG antibodies against whole-inactivated SARS-CoV-2 and mutant S-RBDs as well as virus-neutralizing antibodies in plasma. The development of an animal model to study COVID-19 sex differences will allow for a greater mechanistic understanding of the SARS-CoV-2-associated sex differences seen in the human population. IMPORTANCE Men experience more severe outcomes from coronavirus disease 2019 (COVID-19) than women. Golden Syrian hamsters were used to explore sex differences in the pathogenesis of a human isolate of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). After inoculation, male hamsters experienced greater sickness, developed more severe lung pathology, and recovered more slowly than females. Sex differences in disease could not be reversed by estradiol treatment in males and were not explained by either virus replication kinetics or the concentrations of inflammatory cytokines in the lungs. During the recovery period, antiviral antibody responses in the respiratory tract and plasma, including to newly emerging SARS-CoV-2 variants, were greater in female than in male hamsters. Greater lung pathology during the acute phase combined with lower antiviral antibody responses during the recovery phase of infection in males than in females illustrate the utility of golden Syrian hamsters as a model to explore sex differences in the pathogenesis of SARS-CoV-2 and vaccine-induced immunity and protection.


Subject(s)
Antibodies, Viral/blood , COVID-19/immunology , Lung/pathology , SARS-CoV-2/immunology , Severity of Illness Index , Animals , Antibody Formation/immunology , Cricetinae , Disease Models, Animal , Estradiol/pharmacology , Female , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Interferon-beta/analysis , Lung/diagnostic imaging , Lung/virology , Male , Sex Factors , Spike Glycoprotein, Coronavirus/immunology , Tumor Necrosis Factor-alpha/analysis , Viral Load
2.
STAR Protoc ; 2(2): 100436, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33912845

ABSTRACT

Recent data suggest that cells respond to infection by upregulating the antiviral cytokine interferon-beta (IFN-ß) in a fraction of infected cells. Approaches are thus needed to study these responses on a single-cell level rather than bulk population. Here, we describe a protocol to analyze the IFN-ß response of individual cells using flow cytometry and immunofluorescence microscopy. We show the heterogeneous IFN-ß response to inactivated Sendai virus and human cytomegalovirus, but this protocol can be adapted to other viruses. For complete details on the use and execution of this protocol, please refer to Hare et al. (2020).


Subject(s)
Fluorescent Dyes/metabolism , Interferon-beta , Single Molecule Imaging/methods , Single-Cell Analysis/methods , Telomerase/metabolism , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Dyes/analysis , Humans , Interferon-beta/analysis , Interferon-beta/metabolism , Telomerase/genetics
3.
J Clin Virol ; 114: 37-42, 2019 05.
Article in English | MEDLINE | ID: mdl-30913521

ABSTRACT

BACKGROUND: Experimental data show that type I interferon has a key role in innate immune response against influenza infection. OBJECTIVE: We compared nasal levels of interferon-α2 and ß among inpatients and outpatients with influenza. STUDY DESIGN: Children younger than 5 years of age with influenza-like illness seeking care at the emergency department within the first 72 h of disease onset were prospectively included. Clinical and demographic data and secretions through nasal wash were obtained. Influenza infection was assessed through reverse-transcription polymerase chain reaction and nasal levels of interferon-α2 and ß were measured by enzyme-linked immunosorbent assay. All patients followed until the end of the disease. RESULTS: One hundred patients were included, of which 24 had confirmed influenza infection, and 5 of them were hospitalized. Subtypes A (H3N2) and B were confirmed in 10 and 14 patients, respectively. Seventy-six patients without influenza, including 48% of outpatients, were recruited as controls. All hospitalized patients were significantly younger regardless of influenza status (age <6 months in 59% vs. 23.2%, p < 0.001). All other data were similar among the groups. Comparing median levels of interferon-α2 among children with influenza, levels were significantly higher in outpatients than in hospitalized patients and were 263.2 pg/mL (25-75 interquartile range: 58.3-634) and detectable in only one patient (90 pg/mL), respectively. The levels of interferon-α2 in controls and those of interferon-ß in all groups were not detected. CONCLUSIONS: Higher levels of interferon-α2 in patients with less severe influenza reinforce experimental evidence about the protective role of interferon-α2 against influenza infection.


Subject(s)
Immunity, Innate , Influenza, Human/immunology , Interferon Type I/analysis , Nose/immunology , Respiratory Tract Infections/immunology , Bodily Secretions/virology , Child, Preschool , Cohort Studies , Female , Hospitalization , Humans , Infant , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Inpatients/statistics & numerical data , Interferon Type I/immunology , Interferon alpha-2/analysis , Interferon alpha-2/immunology , Interferon-beta/analysis , Interferon-beta/immunology , Male , Nose/virology , Outpatients/statistics & numerical data , Respiratory Tract Infections/virology
4.
Cell Physiol Biochem ; 44(2): 607-617, 2017.
Article in English | MEDLINE | ID: mdl-29161701

ABSTRACT

BACKGROUND/AIMS: Type I interferon (IFN-1) production and IFN-1 signaling play critical roles in the host antiviral innate immune responses. Although transcription factor Yin Yang 1 (YY1) has been reported to have a dual activator/repressor role during the regulation of interferon beta (IFN-ß) promoter activity, the roles of YY1 in the regulation of upstream signaling pathways leading to IFN-1 induction and IFN-1 signaling during viral infection remain to be elucidated. METHODS: The roles of YY1 in IFN-1 production and IFN-1 signaling were investigated using immunoblotting, real-time PCR, small interfering RNA (siRNA)-mediated YY1 knockdown, YY1 overexpression by transient transfection, and co-immunoprecipitation, using mouse cells. RESULTS: YY1 was shown to interact with STAT1 in the absence of viral infection. Following viral infection, YY1 protein expression levels were decreased. YY1 knockdown led to a considerable downregulation of phosphorylated (p) TBK1 and pIRF3 expressions, while YY1 overexpression significantly upregulated pTBK1 and pIRF3 expression levels and promoted virus-induced IFN-ß production. Additionally, YY1 knockdown led to a significant upregulation of pSTAT1, pSTAT2 and antiviral interferon-stimulated genes, and inhibited viral replication. CONCLUSION: We demonstrated here that YY1 interacts with STAT1 and dynamically regulates the induction of IFN-1 production and activation of IFN-1 signaling in different stages during viral infection.


Subject(s)
Immunity, Innate , YY1 Transcription Factor/metabolism , Animals , Cell Line , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Immunoprecipitation , Interferon Regulatory Factor-3/metabolism , Interferon-beta/analysis , Interferon-beta/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Simplexvirus/physiology , Transfection , Up-Regulation , Vesiculovirus/physiology , YY1 Transcription Factor/antagonists & inhibitors , YY1 Transcription Factor/genetics
5.
Anal Chem ; 88(2): 1138-46, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26641950

ABSTRACT

New and improved methods are required for the enhanced characterization of complex biopharmaceuticals, especially those with charge and glycan heterogeneity. High resolution separation and mass spectrometry (MS) analysis of intact proteoforms can contribute significantly to the characterization of such proteins, many of which are glycoproteins. Here, we report on capillary zone electrophoresis (CZE) coupled via a commercial CESI sheathless interface to an Orbitrap ELITE MS for the intact analysis of recombinant human interferon-ß1 (Avonex, rhIFN-ß1), a biopharmaceutical with complex glycosylation at a single N-linked site. Using a cross-linked polyethylenimine coating, column efficiencies between 350,000 and 450,000 plates were produced, allowing separation based on charge and subtle hydrodynamic volume differences. A total of 138 proteoforms were found, and 55 were quantitated. Charge species due to deamidation and sialylation were separated by CZE. Given the high column efficiency, isobaric positional isomers of a single sialic acid on biantennary glycan antennae were resolved. Further, triantennary isomers (antenna on α(1-3) or α(1-6) arms) were separated and confirmed by exoglycosidase digestion. Proteoforms of the N-terminal cleavage of methionine were detected by precursor molecular weight and top-down ETD and HCD analysis of the reduced protein. Quantitative analysis suggested potential correlations between the methionine loss with the relative amount of the deamidation, as well as the level of deamidation with glycan structure. We demonstrate that high resolution CZE separation of intact glycoprotein species coupled to MS has significant potential for the in-depth characterization and quantitative analysis of biopharmaceutical proteoforms.


Subject(s)
Interferon-beta/analysis , Interferon-beta/chemistry , Biosimilar Pharmaceuticals/analysis , Biosimilar Pharmaceuticals/chemistry , Electrophoresis, Capillary , Humans , Mass Spectrometry
6.
Virology ; 488: 232-41, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26655241

ABSTRACT

To establish a characteristic host response to predict the pathogenicity and tissue tropism of infectious bronchitis viruses (IBV), we investigated innate immune responses (IIR) and apoptosis in chicken embryo kidney cells (CEKC) and tracheal organ cultures (TOC) infected with three IBV strains. Results showed nephropathogenic IBV strains 885 and QX induced greater apoptosis in CEKC than M41, which induced greater apoptosis in TOCs compared to 885 and QX. Elevated IIR is associated with tissue tropism of different IBV strains. Compared to M41, 885 and QX caused greater induction of toll like receptor 3 (TLR3), melanoma differentiation associated protein 5 (MDA5) and interferon beta (IFN-ß) in CEKC. In contrast, M41 infection caused greater expression of these genes than 885 or QX in TOCs. In summary, greater levels of apoptosis and elevated levels of TLR3, MDA5 and IFN-ß expression are associated with increased pathogenicity of IBV strains in renal and tracheal tissues.


Subject(s)
Apoptosis , Coronavirus Infections/veterinary , Immunity, Innate , Infectious bronchitis virus/immunology , Poultry Diseases/pathology , Poultry Diseases/virology , Viral Tropism , Animals , Cell Line , Chickens , Coronavirus Infections/pathology , Coronavirus Infections/virology , DEAD-box RNA Helicases/analysis , Gene Expression Profiling , Infectious bronchitis virus/physiology , Interferon-beta/analysis , Organ Culture Techniques , Toll-Like Receptor 3/analysis , Trachea/pathology , Trachea/virology
7.
Vaccine ; 33(30): 3518-25, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26049004

ABSTRACT

Modified live virus (MLV) vaccines developed to protect against PRRSV circulating in North America (NA) offer limited protection to highly pathogenic (HP) PRRSV strains that are emerging in Asia. MLV vaccines specific to HP-PRRSV strains commercially available in China provide protection to HP-PRRSV; however, the efficacy of these HP-PRRSV vaccines to current circulating NA PRRS viruses has not been reported. The aim of this study is to investigate whether pigs vaccinated with attenuated Chinese HP-PRRSV vaccine (JXA1-R) are protected from infection by NA PRRSV strain NADC-20. We found that pigs vaccinated with JXA1-R were protected from challenges with HV-PRRSV or NADC-20 as shown by fewer days of clinical fever, reduced lung pathology scores, and lower PRRS virus load in the blood. PRRSV-specific antibodies, as measured by IDEXX ELISA, appeared one week after vaccination and virus neutralizing antibodies were detected four weeks post vaccination. Pigs vaccinated with JXA1-R developed broadly neutralizing antibodies with high titers to NADC-20, JXA1-R, and HV-PRRSV. In addition, we also found that IFN-α and IFN-ß occurred at higher levels in the lungs of pigs vaccinated with JXA1-R. Taken together, our studies provide the first evidence that JXA1-R can confer protection in pigs against the heterologous NA PRRSV strain NADC-20.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , China , Interferon-alpha/analysis , Interferon-beta/analysis , Lung/pathology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/pathology , Severity of Illness Index , Swine , Time Factors , Treatment Outcome , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Load , Viral Vaccines/administration & dosage , Viremia
8.
PLoS One ; 9(12): e114874, 2014.
Article in English | MEDLINE | ID: mdl-25506707

ABSTRACT

The production of IFN- I (IFN-α/ß) is one of the earliest and most important host-protective responses. Interferon regulatory factor 3 (IRF3) is a critical transcriptional factor in the IFN-ß signaling pathway. Although significant progress has been achieved in the regulation of IRF3, the process may be more complicated than previously considered. In the present study, heat shock protein 60 (HSP60, HSPD1) was identified as a novel IRF3-interacting protein. Overexpression of HSPD1 facilitated the phosphorylation and dimerization of IRF3 and enhanced IFN-ß induction induced by SeV infection. In contrast, knockdown of endogenous HSPD1 significantly inhibited the signaling pathway. Furthermore, HSPD1 enhanced activation of the IFN-ß promoter mediated by RIG-I, MDA-5, MAVS, TBK1 and IKKε but not IRF3/5D, a mock phosphorylated form of IRF3. The present study indicated that HSPD1 interacted with IRF3 and it contributed to the induction of IFN-ß.


Subject(s)
Chaperonin 60/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Mitochondrial Proteins/metabolism , Protein Interaction Maps , Amino Acid Sequence , Chaperonin 60/analysis , Chaperonin 60/immunology , HEK293 Cells , HeLa Cells , Humans , Interferon Regulatory Factor-3/analysis , Interferon Regulatory Factor-3/immunology , Interferon-beta/analysis , Interferon-beta/immunology , Mitochondrial Proteins/analysis , Mitochondrial Proteins/immunology , Molecular Sequence Data
9.
PLoS One ; 9(7): e101464, 2014.
Article in English | MEDLINE | ID: mdl-24991887

ABSTRACT

Group A Streptococcus (GAS) infections remain a significant health care problem due to high morbidity and mortality associated with GAS diseases, along with their increasing worldwide prevalence. Macrophages play a key role in the control and clearance of GAS infections. Moreover, pro-inflammatory cytokines production and GAS persistence and invasion are related. In this study we investigated the correlation between the GAS clinical isolates genotypes, their known clinical history, and their ability to modulate innate immune response. We constituted a collection of 40 independent GAS isolates representative of the emm types currently prevalent in France and responsible for invasive (57.5%) and non-invasive (42.5%) clinical manifestations. We tested phagocytosis and survival in mouse bone marrow-derived macrophages and quantified the pro-inflammatory mediators (IL-6, TNF-α) and type I interferon (INF-ß) production. Invasive emm89 isolates were more phagocytosed than their non-invasive counterparts, and emm89 isolates more than the other isolates. Regarding the survival, differences were observed depending on the isolate emm type, but not between invasive and non-invasive isolates within the same emm type. The level of inflammatory mediators produced was also emm type-dependent and mostly invasiveness status independent. Isolates of the emm1 type were able to induce the highest levels of both pro-inflammatory cytokines, whereas emm89 isolates induced the earliest production of IFN-ß. Finally, even within emm types, there was a variability of the innate immune responses induced, but survival and inflammatory mediator production were not linked.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Fasciitis, Necrotizing/immunology , Streptococcal Infections/immunology , Streptococcus pyogenes/metabolism , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bone Marrow Cells/cytology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fasciitis, Necrotizing/microbiology , Fasciitis, Necrotizing/pathology , Genotype , Humans , Immunity, Innate , Interferon-beta/analysis , Interferon-beta/metabolism , Interleukin-6/analysis , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Phagocytosis , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Streptococcus pyogenes/isolation & purification , Tumor Necrosis Factor-alpha/analysis
10.
J Periodontol ; 85(9): 1259-65, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24579764

ABSTRACT

BACKGROUND: Low-level laser irradiation promotes cell viability and wound healing in periodontal tissue. However, its effect on periodontal pathogenic bacteria is unknown. The purpose of this study is to investigate the biologic effect of low-level laser irradiation on Porphyromonas gingivalis. METHODS: A murine macrophage cell line (RAW 264.7) was cultured and treated with gallium-aluminum-arsenate (GaAlAs) laser-irradiated P. gingivalis with varying levels of energy fluency. Gene expression of monocyte chemotactic protein-1 (MCP-1), interleukin-6 (IL-6), interferon-ß (IFN-ß), and inducible nitric oxide synthase (iNOS) was examined by reverse transcription-polymerase chain reaction. Production of iNOS was determined by Western blot analysis, and nitric oxide (NO) release was assessed using Griess reagent. Flow cytometric analysis was performed to determine the activation of Toll-like receptors (TLRs) in response to P. gingivalis. RESULTS: The laser-irradiated P. gingivalis significantly enhanced messenger RNA and protein levels of iNOS in RAW 264.7. Although the laser irradiation on P. gingivalis did not alter the expression level of MCP-1, IL-6, and IFN-ß, it showed a noticeable effect on NO production in RAW 264.7. Furthermore, the laser-irradiated P. gingivalis accelerated TLR2 activation, but not TLR4 activation. CONCLUSIONS: This study reveals that GaAlAs laser irradiation on P. gingivalis induced iNOS expression at the transcriptional and translation levels and increased NO release in macrophages. Moreover, it is confirmed that this process was mediated specifically by TLR2 activation. These findings suggest that low-level laser irradiation to periodontal pathogenic bacteria could be detrimental to periodontal treatments.


Subject(s)
Lasers, Semiconductor/therapeutic use , Low-Level Light Therapy/methods , Macrophages/microbiology , Nitric Oxide/radiation effects , Porphyromonas gingivalis/radiation effects , Animals , Bacteriological Techniques , CHO Cells , Cell Culture Techniques , Cell Line , Chemokine CCL2/analysis , Cricetulus , Interferon-beta/analysis , Interleukin-6/analysis , Macrophages/metabolism , Mice , Nitric Oxide Synthase Type II/analysis , Porphyromonas gingivalis/metabolism , Toll-Like Receptor 2/analysis , Toll-Like Receptor 4/analysis
11.
J Interferon Cytokine Res ; 34(7): 498-504, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24444338

ABSTRACT

Interferon beta (IFNß) is used as a first-line treatment in relapsing-remitting multiple sclerosis (MS). The occurrence of neutralizing antidrug antibodies (NAbs) against IFNß may reduce treatment response. Therefore, clinical monitoring of NAbs is currently executed using bioassays, but several bioassays are available and it is unclear how well their readouts correlate. We made a comparison between 2 bioassays; myxovirus resistance protein A (MxA) gene expression assay (MGA) and iLite™ anti-Human IFNß bioassay, to measure IFNß-specific NAb titers in 44 MS patients. We further studied how NAb titers affected in vivo transcription of IFN-induced genes myxovirus resistant 1 (MX1) and C-X-C motif chemokine 10 (CXCL10), in addition to serum CXCL10 protein levels. There were significant correlations between NAb titer levels measured with MGA and iLite (Spearman r=0.9368). MX1 and CXCL10 gene expression was strongly induced by IFNß and NAb positivity significantly reduced this expression. A NAb titer of 150 TRU/mL was observed to be a biological cut-point applicable to both assays, since MX1 and CXCL10 expression was greatly reduced or blocked in patients above this titer level. In conclusion, NAb titers measured with the MGA and iLite bioassays are comparable, but the threshold for positivity in both assays does not correspond to the biologically functional cut-point.


Subject(s)
Antibodies, Neutralizing/blood , Biological Assay/standards , Interferon-beta/analysis , Multiple Sclerosis/blood , Biomarkers/metabolism , Chemokine CXCL10/blood , Chemokine CXCL10/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Humans , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Polymerase Chain Reaction , Transcriptome
12.
Mol Immunol ; 57(2): 210-5, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24172224

ABSTRACT

CD14 is an LPS sensing receptor that is primarily expressed in monocytes. CD14 binds and transfers bacterial LPS to the surface TLR4:MD-2 complex to enable its recognition. After recognizing LPS, this complex produces the first intracellular signals via TIRAP and MyD88, after which surface TLR4/LPS complex is rapidly internalized and produces additional signals via TRAM and TRIF. It was recently suggested that CD14 is a key regulator of LPS-induced TLR4 endocytosis and second signaling. In the present study, we showed that surface TLR4 expressions of human primary monocytes and cell line THP-1 were significantly reduced after treatment with anti-CD14 Ab. Among three anti-CD14 Abs with different epitope specificities used in this study, My4, which has an epitope specificity for LPS binding domain of the CD14 molecule, was found to be the most potent at reduction of surface expression of TLR4 as well as CD14. To test the reason for this reduction, we performed an in vitro internalization assay using anti-TLR4 Ab conjugated with toxin. The results of this analysis indicated surface CD14 ligation-mediated TLR4 internalization, and the mechanism of the internalization was found to be partially clathrin-dependent. We next examined NF-κB/AP-1 activation and TNF-α production of THP-1XBlue-CD14 cells in response to LPS challenge with or without My4 pre-treatment. The results revealed that NF-κB/AP-1 activation and TNF-α production of cells treated with My4 were significantly impaired when compared to the control. Our results suggest that membrane CD14 ligation-mediated TLR4 internalization is a novel mechanism for effective down-regulation of surface expression of TLR4 and subsequent reduction of LPS response of human monocytes.


Subject(s)
Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/immunology , Toll-Like Receptor 4/metabolism , Antibodies, Monoclonal/immunology , Cell Line , Clathrin/metabolism , Down-Regulation , Endocytosis , Epitopes/immunology , Humans , Interferon-beta/analysis , Lipopolysaccharide Receptors/metabolism , Monocytes/immunology , Monocytes/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/biosynthesis
13.
Allergy ; 68(7): 870-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23738920

ABSTRACT

BACKGROUND: Exposure to environmental endocrine-disrupting chemicals (EDCs) is associated with allergy, chronic inflammation, and immunodeficiency. Phthalates, the common EDCs used in plastic industry, may act as adjuvants to disrupt immune system and enhance allergy. Plasmacytoid DCs (pDCs) are predominant cells secreting type I interferon (IFN) against infection and are professional antigen-presenting cells in regulating adaptive immunity. However, the effects of phthalates on the function of pDCs are unknown. METHODS: Circulating pDCs were isolated from healthy subjects, were pretreated with diethylhexyl phthalate (DEHP) and butyl benzyl phthalate (BBP), and were stimulated with Toll-like receptor (TLR)-9 agonist CpG. IFN-α/IFN-ß levels, surface markers, and T-cell stimulatory function were investigated using ELISA, flow cytometry, and pDC/T-cell coculture assay. Mechanisms were investigated using receptor antagonists, pathway inhibitors, Western blotting, and chromatin immunoprecipitation. RESULTS: Diethylhexyl phthalate and butyl benzyl phthalate suppressed CpG-induced IFN-α/IFN-ß expression in pDCs, and the effect was reversed by aryl hydrocarbon receptor (AHR) antagonist. Diethylhexyl phthalate suppressed CpG-activated mitogen-activated protein kinase (MAPK)-MEK1/2-ERK-ELK1 and NFκB signaling pathways. Diethylhexyl phthalate suppressed CpG-induced interferon regulatory factor (IRF)-7 expression by suppressing histone H3K4 trimethylation at IRF7 gene promoter region through inhibiting translocation of H3K4-specific trimethyltransferase WDR5 from cytoplasm into nucleus. Butyl benzyl phthalate or diethylhexyl phthalate-treated pDCs suppressed IFN-γ but enhanced IL-13 production by CD4+ T cells. CONCLUSION: Phthalates may interfere with immunity against infection and promote the deviation of Th2 response to increase allergy by acting on human pDCs via suppressing IFN-α/IFN-ß expression and modulating the ability to stimulate T-cell responses.


Subject(s)
Dendritic Cells/drug effects , Epigenomics , Interferon Type I/drug effects , Interferon Type I/genetics , Phthalic Acids/pharmacology , Blotting, Western , Cell Survival , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interferon Type I/metabolism , Interferon-alpha/analysis , Interferon-alpha/immunology , Interferon-alpha/metabolism , Interferon-beta/analysis , Interferon-beta/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Real-Time Polymerase Chain Reaction , Sampling Studies , Sensitivity and Specificity , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
14.
Curr Protoc Immunol ; Chapter 14: Unit14.21.1-11, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21400681

ABSTRACT

The Basic Protocol in this unit describes measurement of murine interferon (IFN)α/ß by intracellular staining for these cytokines and detection by flow cytometry. Alternate protocols detail an enzyme-linked immunoabsorbent assay (ELISA) for IFNα and a biological assay to measure IFN. The FACS assay allows measurement of IFNα/ß production by defined cell populations, while ELISA measures secreted IFNα. The bioassay measures functional antiviral activity and is the most sensitive of the assays discussed. These assays therefore provide complementary methods to assess IFN production by murine cells.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Interferon-alpha/analysis , Interferon-beta/analysis , Animals , Cells, Cultured , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Mice
15.
Anal Bioanal Chem ; 400(1): 295-303, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21318246

ABSTRACT

In this work, the usefulness of capillary electrophoresis-electrospray ionization time-of-flight-mass spectrometry for the analysis of biopharmaceuticals was studied. Noncovalently bound capillary coatings consisting of Polybrene-poly(vinyl sulfonic acid) or Polybrene-dextran sulfate-Polybrene were used to minimize protein and peptide adsorption, and achieve good separation efficiencies. The potential of the capillary electrophoresis-mass spectrometry (CE-MS) system to characterize degradation products was investigated by analyzing samples of the drugs, recombinant human growth hormone (rhGH) and oxytocin, which had been subjected to prolonged storage, heat exposure, and/or different pH values. Modifications could be assigned based on accurate masses as obtained with time-of-flight-mass spectrometry (TOF-MS) and migration times with respect to the parent compound. For heat-exposed rhGH, oxidations, sulfonate formation, and deamidations were observed. Oxytocin showed strong deamidation (up to 40%) upon heat exposure at low pH, whereas at medium and high pH, mainly dimer (>10%) and trisulfide formation (6-7%) occurred. Recombinant human interferon-ß-1a (rhIFN-ß) was used to evaluate the capability of the CE-MS method to assess glycan heterogeneity of pharmaceutical proteins. Analysis of this N-glycosylated protein revealed a cluster of resolved peaks which appeared to be caused by at least ten glycoforms differing merely in sialic acid and hexose N-acetylhexosamine composition. Based on the relative peak area (assuming an equimolar response per glycoform), a quantitative profile could be derived with the disialytated biantennary glycoform as most abundant (52%). Such a profile may be useful for in-process and quality control of rhIFN-ß batches. It is concluded that the separation power provided by combined capillary electrophoresis and TOF-MS allows discrimination of highly related protein species.


Subject(s)
Biological Products/analysis , Electrophoresis, Capillary/methods , Pharmaceutical Preparations/analysis , Spectrometry, Mass, Electrospray Ionization/methods , Growth Hormone/analysis , Humans , Interferon beta-1a , Interferon-beta/analysis , Oxytocin/analysis , Recombinant Proteins/analysis
16.
Eur J Immunol ; 41(1): 224-34, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21182093

ABSTRACT

The inhibitor of κB kinase ε (IKKε) is pivotal for an efficient innate immune response to viral infections and has been recognized as breast cancer oncogene. The antiviral function of IKKε involves activation of the transcription factors IFN regulatory factor 3 (IRF3) and NF-κB, thus inducing the expression of type I IFN. Here, we have identified two novel splice variants of human IKKε, designated IKKε-sv1 and IKKε-sv2, respectively. Interestingly, RT-PCR revealed quantitatively different isoform expression in PBMC from different individuals. Moreover, we found cell type- and stimulus-specific protein expression of the various splice variants. Overexpression of full-length wt IKKε (IKKε-wt) leads to the activation of NF-κB- as well as IRF3-driven luciferase reporter genes. Although none of the splice variants activates IRF3, IKKε-sv1 still activates NF-κB, whereas IKKε-sv2 is also defective in NF-κB activation. Both splice variants form dimers with IKKε-wt and inhibit IKKε-wt-induced IRF3 signaling including the antiviral activity in a dominant-negative manner. The lack of IRF3 activation is likely caused by the failure of the splice variants to interact with the adapter proteins TANK, NAP1, and/or SINTBAD. Taken together, our data suggest alternative splicing as a novel regulatory mechanism suitable to shift the balance between different functions of IKKε.


Subject(s)
Alternative Splicing , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/metabolism , Leukocytes, Mononuclear/immunology , NF-kappa B/metabolism , Virus Diseases/immunology , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Cell Line , Down-Regulation , Humans , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon-beta/analysis , Molecular Sequence Data , Proteins/metabolism , tRNA Methyltransferases
17.
J Immunol Methods ; 350(1-2): 106-17, 2009 Oct 31.
Article in English | MEDLINE | ID: mdl-19666024

ABSTRACT

This is a flow cytometric study of expression of a diverse set of myeloid receptors on murine splenic plasmacytoid dendritic cells (pDCs) and the description of a FACS based assay for measurement of interferon (IFN)alpha/beta. We have extended the known repertoire of PRR expressed on murine pDCs with the novel observation that they express Dectin-2 and contain intracellular MR. In addition, this is the first report of F4/80 and CD200 on murine pDCs. We have confirmed the observation by others that murine pDCs express CD200R, the lectin Dectin-1 and the scavenger receptor CD36. This report also details a flow cytometry-based protocol to measure the production of murine IFNalpha/beta by splenic pDC. Briefly, splenocytes can be stimulated with virus or a TLR9 agonist and IFNalpha/beta production by pDCs is detected following intracellular staining. pDCs are specifically identified by 120G8 staining at 6 h after stimulation with inactivated influenza virus, however the specificity of 120G8 for pDCs is reduced at times later than 12 h. This assay is suitable for use with splenocytes from some mouse strains (129/SvEv), but not others (C57BL/6J), probably due to C57BL6J producing insufficient amounts of IFN following stimulation to be detected by intracellular staining. However, IFN production by C57BL/6J splenocytes is readily detectable by bioassay. In addition to being more sensitive than intracellular staining, the bioassay is also more sensitive than an IFNalpha ELISA. The comparable sensitivities of these assays are often a critical determinant of the choice of assay and are an important consideration in experimental design.


Subject(s)
Antigens, CD/analysis , Antigens, Differentiation/analysis , Dendritic Cells/metabolism , Flow Cytometry/methods , Interferon-alpha/analysis , Interferon-beta/analysis , Lectins, C-Type/analysis , Plasma Cells/metabolism , Animals , Antigens, CD/biosynthesis , Antigens, CD/immunology , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/immunology , CD36 Antigens/analysis , CD36 Antigens/biosynthesis , CD36 Antigens/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/physiology , Interferon-alpha/biosynthesis , Interferon-alpha/immunology , Interferon-beta/biosynthesis , Interferon-beta/immunology , Lectins, C-Type/biosynthesis , Lectins, C-Type/immunology , Mice , Mice, Knockout , Plasma Cells/cytology , Plasma Cells/immunology , Sensitivity and Specificity
18.
Anal Chem ; 80(19): 7473-81, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18729476

ABSTRACT

Unlike small-molecule drugs, the conformational properties of protein biopharmaceuticals in solution are influenced by a variety of factors that are not solely defined by their covalent chemical structure. Since the conformation (or higher order structure) of a protein is a major modulator of its biological activity, the ability to detect changes in both the higher order structure and conformational dynamics of a protein, induced by an array of extrinsic factors, is of central importance in producing, purifying, and formulating a commercial biopharmaceutical with consistent therapeutic properties. In this study we demonstrate that two complementary mass spectrometry-based approaches (analysis of ionic charge-state distribution and hydrogen/deuterium exchange) can be a potent tool in monitoring conformational changes in protein biopharmaceuticals. The utility of these approaches is demonstrated by detecting and characterizing conformational changes in the biopharmaceutical product interferon beta-1a (IFN-beta-1a). The protein degradation process was modeled by inducing a single chemical modification of IFN-beta1a (alkylation of its only free cysteine residue with N-ethylmaleimide), which causes significant reduction in its antiviral activity. Analysis of IFN-beta1a ionic charge-state distributions unequivocally reveals a significant decrease of conformational stability in the degraded protein, while hydrogen/deuterium exchange measurements provide a clear indication that the higher order structure is affected well beyond the covalent modification site. Importantly, neither technique required that the location or indeed the nature of the chemical modification be known prior to or elucidated in the process of the analysis. In contrast, application of the standard armamentarium of biophysical tools, which are commonly employed for quality control of protein pharmaceuticals, met with very limited success in detection and characterization of conformational changes in the modified IFN-beta1a. This work highlights the role mass spectrometry can and should play in the biopharmaceutical industry beyond the presently assigned task of primary structure analysis.


Subject(s)
Interferon-beta/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Amino Acid Sequence , Cardiovirus Infections/drug therapy , Cardiovirus Infections/virology , Cell Line, Tumor , Cytopathogenic Effect, Viral/drug effects , Encephalomyocarditis virus/physiology , Ethylmaleimide/analogs & derivatives , Ethylmaleimide/chemistry , Humans , Interferon beta-1a , Interferon-beta/analysis , Interferon-beta/pharmacology , Lung/drug effects , Lung/virology , Models, Molecular , Molecular Sequence Data , Protein Conformation , Recombinant Proteins/analysis , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
19.
Brain ; 131(Pt 6): 1455-63, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18474519

ABSTRACT

B cells are increasingly recognized as major players in multiple sclerosis pathogenesis. The BAFF/APRIL system is crucial for B cell homoeostasis and may drive B cell-dependent autoimmunity. We asked whether this system is affected by Interferon (IFN)-beta therapy. We analysed transcription of the ligands (BAFF, APRIL, TWE-PRIL) and the corresponding receptors (BAFF-R, TACI and BCMA) by TaqMan-PCR ex vivo in whole blood and in immune cell subsets purified from IFN-beta-treated multiple sclerosis patients. Serum BAFF concentrations were determined by ELISA. This cross-sectional study involved 107 donors. IFN-beta therapy strongly induced BAFF transcription proportionally to the IFN-beta biomarker MxA in monocytes and granulocytes in vivo. BAFF serum concentrations were elevated in IFN-beta-treated multiple sclerosis patients to a similar level as observed in SLE patients. In cultured PBMC, neutrophils, fibroblasts and astrocytes, BAFF was induced by IFN-beta concentrations similar to those reached in vivo in treated multiple sclerosis patients. BAFF turned out to be the main regulated element of the BAFF/APRIL system. In untreated multiple sclerosis patients, there was no BAFF increase as compared to healthy controls. Our study reveals a complex situation. We show that IFN-beta therapy induces a potent B cell survival factor, BAFF. However, B cell depletion would be desirable at least in some multiple sclerosis patients. The systemic induction of BAFF by IFN-beta therapy may facilitate the production of various autoantibodies and of IFN-neutralizing antibodies. Individual MS/NMO patients who have major B cell involvement may benefit less than others from IFN-beta therapy, thus explaining interindividual differences of the therapeutic response.


Subject(s)
B-Cell Activating Factor/metabolism , B-Lymphocytes/drug effects , Immunotherapy/methods , Interferon-beta/therapeutic use , Multiple Sclerosis/drug therapy , Adult , Autoimmunity , B-Cell Activating Factor/blood , B-Cell Activation Factor Receptor/metabolism , B-Lymphocytes/immunology , Case-Control Studies , Cells, Cultured , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Interferon-beta/analysis , Male , Multiple Sclerosis/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
20.
J Gen Virol ; 88(Pt 9): 2559-2567, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17698667

ABSTRACT

The matrix (M) protein of vesicular stomatitis virus (VSV) is a multi-functional protein involved in virus assembly, budding and pathogenesis. The (24)PPPY(27) late (L) domain of the M protein plays a key role in virus budding, whereas amino acids downstream of the PPPY motif contribute to host protein shut-off and pathogenesis. Using a panel of (37)PSAP(40) recombinant viruses, it has been demonstrated previously that the PSAP region of M does not possess L-domain activity similar to that of PPPY in BHK-21 cells. This study reports the unanticipated finding that these PSAP recombinants were attenuated in cell culture and in mice compared with control viruses. Indeed, PSAP recombinant viruses exhibited a small-plaque phenotype, reduced CPE, reduced levels of activated caspase-3, enhanced production of IFN-beta and reduced titres in the lungs and brains of infected mice. In particular, recombinant virus M6PY>A4-R34E was the most severely attenuated, exhibiting little or no CPE in cell culture and undetectable titres in the lungs and brains of infected mice. These findings indicate an important role for the PSAP region (aa 33-44) of the M protein in the pathology of VSV infection and may have implications for the development of VSV as a vaccine and/or oncolytic vector.


Subject(s)
Vesicular stomatitis Indiana virus/genetics , Viral Matrix Proteins/physiology , Amino Acid Sequence , Animals , Caspase 3/metabolism , Cell Adhesion , Cricetinae , HeLa Cells , Humans , Interferon-beta/analysis , Kidney , Mice , Recombination, Genetic , Rhabdoviridae Infections/immunology , Viral Matrix Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...