Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 490(4): 1301-1306, 2017 09 02.
Article in English | MEDLINE | ID: mdl-28688767

ABSTRACT

The interplay between highly pathogenic avian influenza (HPAI) H5N1 virus and immune cells has been extensively studied for years, as host immune components are thought to play significant roles in promoting the systemic spread of the virus and responsible for cytokine storm. Previous studies suggested that the interaction of B cells and monocytes could promote HPAI H5N1 infection by enhancing avian influenza virus receptor expression. In this study, we further investigate the relationship between the HPAI H5N1 virus, activated B cells, and DC-SIGN expression. DC-SIGN has been described as an important factor for mediating various types of viral infection. Here, we first demonstrate that HPAI H5N1 infection could induce an activation of B cells, which was associated with DC-SIGN expression. Using CD40L and recombinant IL-4 for B cell stimulation, we determined that DC-SIGN expressed on activated B cells was able to enhance its susceptibility to HPAI H5N1 infection. Our findings uncover the interplay between this H5N1 virus and B cells and provide important information in understanding how the virus overcomes our immune system, contributing to its unusual immunopathogenesis.


Subject(s)
B-Lymphocytes/virology , Cell Adhesion Molecules/immunology , Host-Pathogen Interactions , Influenza A Virus, H5N1 Subtype/physiology , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , Animals , B-Lymphocytes/immunology , B7-2 Antigen/genetics , B7-2 Antigen/immunology , Birds/virology , CD40 Ligand/pharmacology , Cell Adhesion Molecules/genetics , Disease Susceptibility , Gene Expression Regulation , Humans , Influenza A Virus, H5N1 Subtype/isolation & purification , Interleukin-4/pharmacology , Lectins, C-Type/genetics , Lymphocyte Activation/drug effects , Primary Cell Culture , Receptors, Cell Surface/genetics , Recombinant Proteins/pharmacology , Signal Transduction
2.
J Biomater Sci Polym Ed ; 28(15): 1786-1795, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28571525

ABSTRACT

Antibodies are commonly used in diagnostic routines to identify pathogens. The testing protocols are relatively simple, requiring a certain amount of a specific antibody to detect its corresponding pathogen. Antibody functionality can be mimicked by synthesizing molecularly imprinted polymers (MIPs), i.e. polymers that can selectively recognize a given template structure. Thus, MIPs are sometimes termed 'plastic antibody (PA)'. In this study, we have synthesized new granular MIPs using influenza A virus templates by precipitation polymerization. The selective binding of influenza A to the MIP particles was assessed and subsequently contrasted with other viruses. The affinities of influenza A virus towards the MIP was estimated based on an agglutination test by measuring the amount of influenza subtypes absorbed onto the MIPs. The MIPs produced using the H1N1 template showed specific reactivity to H1N1 while those produced using H5N1 and H3N2 templates showed cross-reactivity.


Subject(s)
Agglutination Tests , Antibodies/metabolism , Influenza A virus/metabolism , Molecular Imprinting , Polymers/chemical synthesis , Polymers/metabolism , Particle Size , Polymers/chemistry
3.
PLoS One ; 12(5): e0176974, 2017.
Article in English | MEDLINE | ID: mdl-28494003

ABSTRACT

One of the key pathophysiologies of H5N1 infection is excessive proinflammatory cytokine response (cytokine storm) characterized by increases in IFN-ß, TNF-α, IL-6, CXCL10, CCL4, CCL2 and CCL5 in the respiratory tract. H5N1-induced cytokine release can occur via an infection-independent mechanism, however, detail of the cellular signaling involved is poorly understood. To elucidate this mechanism, the effect of inactivated (ß-propiolactone-treated) H5N1 on the cytokine and chemokine mRNA expression in 16HBE14o- human respiratory epithelial cells was investigated. We found that the inactivated-H5N1 increased mRNA for IL-6 and CXCL8 but not TNF-α, CCL5 or CXCL10. This effect of the inactivated-H5N1 was inhibited by sialic acid receptor inhibitor (α-2,3 sialidase), adenosine diphosphatase (apyrase), P2Y receptor (P2YR) inhibitor (suramin), P2Y6R antagonist (MRS2578), phospholipase C inhibitor (U73122), protein kinase C inhibitors (BIM and Gö6976) and cell-permeant Ca2+ chelator (BAPTA-AM). Inhibitors of MAPK signaling, including of ERK1/2 (PD98059), p38 MAPK (SB203580) and JNK (SP600125) significantly suppressed the inactivated-H5N1-induced mRNA expression of CXCL8. On the other hand, the inactivated-H5N1-induced mRNA expression of IL-6 was inhibited by SB203580, but not PD98059 or SP600125, whereas SN-50, an inhibitor of NF-κB, inhibited the effect of virus on mRNA expression of both of IL-6 and CXCL8. Taken together, our data suggest that, without infection, inactivated-H5N1 induces mRNA expression of IL-6 and CXCL8 by a mechanism, or mechanisms, requiring interaction between viral hemagglutinin and α-2,3 sialic acid receptors at the cell membrane of host cells, and involves activation of P2Y6 purinergic receptors.


Subject(s)
Gene Expression Regulation , Influenza A Virus, H5N1 Subtype/physiology , Influenza, Human/genetics , Interleukin-6/genetics , Interleukin-8/genetics , Receptors, Purinergic P2/metabolism , Respiratory Mucosa/virology , Animals , Cell Line , Chickens , Humans , Influenza, Human/metabolism , Influenza, Human/virology , RNA, Messenger/genetics , Respiratory Mucosa/metabolism , Signal Transduction
4.
J Immunol ; 195(9): 4378-86, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26408671

ABSTRACT

Studies of influenza-specific immune responses in humans have largely assessed systemic responses involving serum Ab and peripheral blood T cell responses. However, recent evidence indicates that tissue-resident memory T (TRM) cells play an important role in local murine intrapulmonary immunity. Rhesus monkeys were pulmonary exposed to 2009 pandemic H1N1 virus at days 0 and 28 and immune responses in different tissue compartments were measured. All animals were asymptomatic postinfection. Although only minimal memory immune responses were detected in peripheral blood, a high frequency of influenza nucleoprotein-specific memory T cells was detected in the lung at the "contraction phase," 49-58 d after second virus inoculation. A substantial proportion of lung nucleoprotein-specific memory CD8(+) T cells expressed CD103 and CD69, phenotypic markers of TRM cells. Lung CD103(+) and CD103(-) memory CD8(+) T cells expressed similar levels of IFN-γ and IL-2. Unlike memory T cells, spontaneous Ab secreting cells and memory B cells specific to influenza hemagglutinin were primarily observed in the mediastinal lymph nodes. Little difference in systemic and local immune responses against influenza was observed between young adult (6-8 y) and old animals (18-28 y). Using a nonhuman primate model, we revealed substantial induction of local T and B cell responses following 2009 pandemic H1N1 infection. Our study identified a subset of influenza-specific lung memory T cells characterized as TRM cells in rhesus monkeys. The rhesus monkey model may be useful to explore the role of TRM cells in local tissue protective immunity after rechallenge and vaccination.


Subject(s)
B-Lymphocytes/immunology , Immunologic Memory/immunology , Influenza A Virus, H1N1 Subtype/immunology , Macaca mulatta/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes/immunology , Age Factors , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Host-Pathogen Interactions/immunology , Humans , Influenza A Virus, H1N1 Subtype/physiology , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/virology , Macaca mulatta/metabolism , Macaca mulatta/virology , Mediastinum/virology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Spleen/immunology , Spleen/metabolism , Spleen/virology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Time Factors
5.
Biochem Biophys Res Commun ; 464(3): 888-93, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26187669

ABSTRACT

The highly pathogenic avian influenza (HPAI) H5N1 virus causes severe systemic infection in avian and mammalian species, including humans by first targeting immune cells. This subsequently renders the innate and adaptive immune responses less active, thus allowing dissemination of the virus to systemic organs. To gain insight into the pathogenesis of H5N1, this study aims to determine the susceptibility of human PBMCs to the H5N1 virus and explore the factors which influence this susceptibility. We found that PBMCs were a target of H5N1 infection, and that monocytes and B cells were populations which were clearly the most susceptible. Analysis of PBMC subpopulations showed that isolated monocytes and monocytes residing in whole PBMCs had comparable percentages of infection (28.97 ± 5.54% vs 22.23 ± 5.14%). In contrast, isolated B cells were infected to a much lower degree than B cells residing in a mixture of whole PBMCs (0.88 ± 0.34% vs 34.87 ± 4.63%). Different susceptibility levels of B cells for these tested conditions spurred us to explore the B cell-H5N1 interaction mechanisms. Here, we first demonstrated that monocytes play a crucial role in the enhancement of B cell susceptibility to H5N1 infection. Although the actual mechanism by which this enhancement occurs remains in question, α2,3-linked sialic acid (SA), known for influenza virus receptors, could be a responsible factor for the greater susceptibility of B cells, as it was highly expressed on the surface of B cells upon H5N1 infection of B cell/monocyte co-cultures. Our findings reveal some of the factors involved with the permissiveness of human immune cells to H5N1 virus and provide a better understanding of the tropism of H5N1 in immune cells.


Subject(s)
B-Lymphocytes/virology , Influenza A Virus, H5N1 Subtype/pathogenicity , Monocytes/virology , Receptors, Cell Surface/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Coculture Techniques , Disease Susceptibility , Humans , Influenza, Human/virology , Leukocytes, Mononuclear/virology , Monocytes/immunology , Up-Regulation
6.
Am J Trop Med Hyg ; 90(1): 149-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24277784

ABSTRACT

We studied cross-reactive antibodies against avian influenza H5N1 and 2009 pandemic (p) H1N1 in 200 serum samples from US military personnel collected before the H1N1 pandemic. Assays used to measure antibodies against viral proteins involved in protection included a hemagglutination inhibition (HI) assay and a neuraminidase inhibition (NI) assay. Viral neutralization by antibodies against avian influenza H5N1 and 2009 pH1N1 was assessed by influenza (H5) pseudotyped lentiviral particle-based and H1N1 microneutralization assays. Some US military personnel had cross-neutralizing antibodies against H5N1 (14%) and 2009 pH1N1 (16.5%). The odds of having cross-neutralizing antibodies against 2009 pH1N1 were 4.4 times higher in subjects receiving more than five inactivated whole influenza virus vaccinations than those subjects with no record of vaccination. Although unclear if the result of prior vaccination or disease exposure, these pre-existing antibodies may prevent or reduce disease severity.


Subject(s)
Antibodies, Viral/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza, Human/virology , Military Personnel , Adult , Aged , Antibodies, Neutralizing , Female , Humans , Influenza, Human/blood , Influenza, Human/immunology , Male , Middle Aged , Neutralization Tests , Odds Ratio , United States
7.
PLoS One ; 8(12): e81805, 2013.
Article in English | MEDLINE | ID: mdl-24339969

ABSTRACT

The presence of abnormal hematologic findings such as lymphopenia, thrombocytopenia, and pancytopenia were diagnosed in severe cases of avian influenza A H5N1. Whether direct viral dissemination to bone marrow (BM) cells causes this phenomenon remains elusive. We explore the susceptibility of the two stem cell types; hematopoietic stem cells (HSCs) and mesenchymal stromal cells (MSCs) isolated from human BM cells or cord blood, to infection with avian H5N1 viruses. For the first time, we demonstrated that the H5N1 virus could productively infect and induce cell death in both human stem cell types. In contrast, these activities were not observed upon human influenza virus infection. We also determined whether infection affects the immunomodulatory function of MSCs. We noted a consequent dysregulation of MSC-mediated immune modulation as observed by high cytokine and chemokine production in H5N1 infected MSCs and monocytes cocultures. These findings provide a better understanding of H5N1 pathogenesis in terms of broad tissue tropism and systemic spread.


Subject(s)
Antigens, CD34 , Hematopoietic Stem Cells/virology , Influenza A Virus, H5N1 Subtype/physiology , Influenza, Human/metabolism , Mesenchymal Stem Cells/virology , Viral Tropism/physiology , Female , Fetal Blood/metabolism , Fetal Blood/virology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Influenza, Human/pathology , Influenza, Human/therapy , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology
8.
PLoS One ; 8(3): e59674, 2013.
Article in English | MEDLINE | ID: mdl-23555741

ABSTRACT

INTRODUCTION: Recent studies have demonstrated that inactivated seasonal influenza vaccines (IIV) may elicit production of heterosubtypic antibodies, which can neutralize avian H5N1 virus in a small proportion of subjects. We hypothesized that prime boost regimens of live and inactivated trivalent seasonal influenza vaccines (LAIV and IIV) would enhance production of heterosubtypic immunity and provide evidence of cross-protection against other influenza viruses. METHODS: In an open-label study, 26 adult volunteers were randomized to receive one of four vaccine regimens containing two doses of 2009-10 seasonal influenza vaccines administered 8 (±1) weeks apart: 2 doses of LAIV; 2 doses of IIV; LAIV then IIV; IIV then LAIV. Humoral immunity assays for avian H5N1, 2009 pandemic H1N1 (pH1N1), and seasonal vaccine strains were performed on blood collected pre-vaccine and 2 and 4 weeks later. The percentage of cytokine-producing T-cells was compared with baseline 14 days after each dose. RESULTS: Subjects receiving IIV had prompt serological responses to vaccine strains. Two subjects receiving heterologous prime boost regimens had enhanced haemagglutination inhibition (HI) and neutralization (NT) titres against pH1N1, and one subject against avian H5N1; all three had pre-existing cross-reactive antibodies detected at baseline. Significantly elevated titres to H5N1 and pH1N1 by neuraminidase inhibition (NI) assay were observed following LAIV-IIV administration. Both vaccines elicited cross-reactive CD4+ T-cell responses to nucleoprotein of avian H5N1 and pH1N1. All regimens were safe and well tolerated. CONCLUSION: Neither homologous nor heterologous prime boost immunization enhanced serum HI and NT titres to 2009 pH1N1 or avian H5N1 compared to single dose vaccine. However heterologous prime-boost vaccination did lead to in vitro evidence of cross-reactivity by NI; the significance of this finding is unclear. These data support the strategy of administering single dose trivalent seasonal influenza vaccine at the outset of an influenza pandemic while a specific vaccine is being developed. TRIAL REGISTRATION: ClinicalTrials.gov NCT01044095.


Subject(s)
Cross Reactions , Immunization, Secondary/methods , Influenza in Birds/immunology , Influenza, Human/prevention & control , Orthomyxoviridae/immunology , Pandemics/prevention & control , Vaccination/methods , Adolescent , Adult , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Birds , Feasibility Studies , Female , Health , Humans , Immunization, Secondary/adverse effects , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/physiology , Influenza in Birds/prevention & control , Influenza, Human/epidemiology , Influenza, Human/immunology , Male , Middle Aged , Orthomyxoviridae/physiology , Pilot Projects , Safety , Seasons , T-Lymphocytes/immunology , T-Lymphocytes/virology , Vaccination/adverse effects , Viral Vaccines/adverse effects , Viral Vaccines/immunology , Young Adult
9.
Biochem Biophys Res Commun ; 398(4): 752-8, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20627090

ABSTRACT

Information on the immune response against H5N1 within the lung is lacking. Here we describe the sustained antiviral immune responses, as indicated by the expression of MxA protein and IFN-alpha mRNA, in autopsy lung tissue from an H5N1-infected patient. H5N1 infection of primary bronchial/tracheal epithelial cells and lung microvascular endothelial cells induced IP-10, and also up-regulated the retinoic acid-inducible gene-I (RIG-I). Down-regulation of RIG-I gene expression decreased IP-10 response. Co-culturing of H5N1-infected pulmonary cells with TNF-alpha led to synergistically enhanced production of IP-10. In the absence of viral infection, TNF-alpha and IFN-alpha also synergistically enhanced IP-10 response. Methylprednisolone showed only a partial inhibitory effect on this chemokine response. Our findings strongly suggest that both the H5N1 virus and the locally produced antiviral cytokines; IFN-alpha and TNF-alpha may have an important role in inducing IP-10 hyperresponse, leading to inflammatory damage in infected lung.


Subject(s)
Chemokine CXCL10/biosynthesis , Influenza A Virus, H5N1 Subtype , Influenza, Human/immunology , Lung/immunology , Lung/virology , Pneumonia, Viral/immunology , Cells, Cultured , Chemokine CXCL10/antagonists & inhibitors , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , GTP-Binding Proteins/biosynthesis , Humans , Interferon-alpha/biosynthesis , Interferon-alpha/pharmacology , Methylprednisolone/pharmacology , Myxovirus Resistance Proteins , Receptors, Immunologic , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
J Immunol ; 179(8): 5220-7, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17911607

ABSTRACT

There is worldwide concern that the avian influenza H5N1 virus, with a mortality rate of >50%, might cause the next influenza pandemic. Unlike most other influenza infections, H5N1 infection causes a systemic disease. The underlying mechanisms for this effect are still unclear. In this study, we investigate the interplay between avian influenza H5N1 and human dendritic cells (DC). We showed that H5N1 virus can infect and replicate in monocyte-derived and blood myeloid DC, leading to cell death. These results suggest that H5N1 escapes viral-specific immunity, and could disseminate via DC. In contrast, blood pDC were resistant to infection and produced high amounts of IFN-alpha. Addition of this cytokine to monocyte-derived DC or pretreatment with TLR ligands protected against infection and the cytopathic effects of H5N1 virus.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Disease Susceptibility/immunology , Influenza A Virus, H5N1 Subtype/immunology , Interferon-alpha/metabolism , Toll-Like Receptors/metabolism , Animals , Antiviral Agents/metabolism , Birds , Cell Lineage/immunology , Cells, Cultured , Coculture Techniques , Cytopathogenic Effect, Viral/immunology , Dendritic Cells/metabolism , Humans , Immunity, Innate , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/immunology , Influenza in Birds/metabolism , Influenza in Birds/prevention & control , Interferon-alpha/physiology , Ligands , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Molecular Sequence Data , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/physiology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/physiology , Toll-Like Receptors/physiology , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...