Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
2.
Curr Trends Immunol ; 17: 117-123, 2016.
Article in English | MEDLINE | ID: mdl-28133423

ABSTRACT

Promising drugs to treat Ebola virus (EBOV) infection are currently being developed, but so far none has shown efficacy in clinical trials. Drugs that can stimulate host innate defense responses may retard the progression of EBOV disease. We report here the dramatic effect of hemin, the natural inducer of the heme catabolic enzyme heme oxygenase-1 (HO-1), in the reduction of EBOV replication. Treatment of primary monocyte-derived macrophages (MDM), Vero E6 cells, HeLa cells, and human foreskin fibroblasts (HFF1) with hemin reduced EBOV infection by >90%, and showed minimal toxicity to infected cells. Inhibition of HO-1 enzymatic activity and silencing HO-1 expression prevented the hemin-mediated suppression of EBOV infection, suggesting an important role for induction of this intracellular mediator in restricting EBOV replication. The inverse correlation between hemin-induced HO-1 and EBOV replication provides a potentially useful therapeutic modality based on the stimulation of an innate cellular response against Ebola infection.

3.
J Biol Chem ; 290(7): 4422-31, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25548275

ABSTRACT

Pregnancy-specific glycoproteins (PSGs) are a family of Ig-like proteins secreted by specialized placental cells. The PSG1 structure is composed of a single Ig variable region-like N-terminal domain and three Ig constant region-like domains termed A1, A2, and B2. Members of the human and murine PSG family have been shown to induce anti-inflammatory cytokines from monocytes and macrophages and to stimulate angiogenesis. We recently showed that recombinant forms of PSG1 (PSG1-Fc and PSG1-His) and PSG1 purified from the serum of pregnant women are associated with the immunoregulatory cytokine TGF-ß1 and activated latent TGF-ß1. Here, we sought to examine the requirement of specific PSG1 domains in the activation of latent TGF-ß1. Plasmon surface resonance studies showed that PSG1 directly bound to the small latent complex and to the latency-associated peptide of TGF-ß1 and that this binding was mediated through the B2 domain. Furthermore, the B2 domain alone was sufficient for activating the small latent complex. In separate experiments, we found that the PSG1-mediated induction of TGF-ß1 secretion in macrophages was dependent on the N-terminal domain. Mutagenesis analysis revealed that four amino acids (LYHY) of the CC' loop of the N-terminal domain were required for induction of latent TGF-ß1 secretion. Together, our results show that two distinct domains of PSG1 are involved in the regulation of TGF-ß1 and provide a mechanistic framework for how PSGs modulate the immunoregulatory environment at the maternal-fetal interface for successful pregnancy outcome.


Subject(s)
Macrophages/metabolism , Monocytes/metabolism , Placenta/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Blotting, Western , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Immunoenzyme Techniques , Macrophages/cytology , Mice , Monocytes/cytology , Placenta/cytology , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/genetics , Protein Conformation , Protein Structure, Tertiary , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/genetics
4.
Antiviral Res ; 98(2): 325-31, 2013 May.
Article in English | MEDLINE | ID: mdl-23528258

ABSTRACT

Three different antiviral assays were developed for the in vitro screening of inhibitors of the hepatitis A virus (HAV) of which (i) a cytopathic effect reduction assay suitable for medium-to-high-throughput screening and (ii) two virus yield reduction assays (based on quantification of viral RNA) for genotypes IB and IIIA. The assays were validated for antiviral studies with interferon-alpha (IFNα) and amantadine HCl, two known inhibitors of HAV replication. IFNα effectively inhibited HAV replication, whereas the activity of amantadine HCl appeared to be strain-dependent. Employing these assays, we assessed the effect of the known enterovirus inhibitors pleconaril, rupintrivir and enviroxime on HAV replication. Pleconaril exhibited some very moderate activity, the effect of rupintrivir proved to be strain-dependent. Enviroxime did not inhibit HAV replication, suggesting that phosphatidylinositol-4-kinase IIIß is not crucial in the HAV life cycle.


Subject(s)
Antiviral Agents/pharmacology , Biological Assay/methods , Hepatitis A virus/drug effects , Hepatitis A virus/physiology , Hepatitis A/virology , Virus Replication/drug effects , Cell Line , Cytopathogenic Effect, Viral , Hepatitis A virus/genetics , Humans
5.
Gastroenterology ; 142(7): 1516-25.e3, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22430395

ABSTRACT

BACKGROUND & AIMS: CD4+ T-regulatory (Treg) cells suppress immune responses and control self-tolerance and immunity to pathogens, cancer, and alloantigens. Most pathogens activate Treg cells to minimize immune-mediated tissue damage and prevent clearance, which promotes chronic infections. However, hepatitis A virus (HAV) temporarily inhibits Treg-cell functions. We investigated whether the interaction of HAV with its cellular receptor 1 (HAVCR1), a T-cell co-stimulatory molecule, inhibits the function of Treg cells to control HAV infection. METHODS: We studied the effects of HAV interaction with HAVCR1 on human T cells using binding, signal transduction, apoptosis, activation, suppression, cytokine production, and confocal microscopy analyses. Cytokines were analyzed in sera from 14 patients with HAV infection using bead arrays. RESULTS: Human Treg cells constitutively express HAVCR1. Binding of HAV to HAVCR1 blocked phosphorylation of Akt, prevented activation of the T-cell receptor, and inhibited function of Treg cells. At the peak viremia, patients with acute HAV infection had no Treg-cell suppression function, produced low levels of transforming growth factor-ß , which limited leukocyte recruitment and survival, and produced high levels of interleukin-22, which prevented liver damage. CONCLUSIONS: Interaction between HAV and its receptor HAVCR1 inhibits Treg-cell function, resulting in an immune imbalance that allows viral expansion with limited hepatocellular damage during early stages of infection-a characteristic of HAV pathogenesis. The mechanism by which HAV is cleared in the absence of Treg-cell function could be used as a model to develop anticancer therapies, modulate autoimmune and allergic responses, and prevent transplant rejection.


Subject(s)
Hepatitis A virus/metabolism , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , T-Lymphocytes, Regulatory/immunology , Virus Attachment , Cell Line , Hepatitis A/immunology , Hepatitis A/metabolism , Hepatitis A Virus Cellular Receptor 1 , Humans , Interleukins/biosynthesis , Proto-Oncogene Proteins c-akt , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Transforming Growth Factor beta1/blood , Interleukin-22
6.
Vaccine ; 29(16): 2968-77, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21329775

ABSTRACT

Ebola virus is a Filoviridae that causes hemorrhagic fever in humans and induces high morbidity and mortality rates. Filoviruses are classified as "Category A bioterrorism agents", and currently there are no licensed therapeutics or vaccines to treat and prevent infection. The Filovirus glycoprotein (GP) is sufficient to protect individuals against infection, and several vaccines based on GP are under development including recombinant adenovirus, parainfluenza virus, Venezuelan equine encephalitis virus, vesicular stomatitis virus (VSV) and virus-like particles. Here we describe the development of a GP Fc fusion protein as a vaccine candidate. We expressed the extracellular domain of the Zaire Ebola virus (ZEBOV) GP fused to the Fc fragment of human IgG1 (ZEBOVGP-Fc) in mammalian cells and showed that GP undergoes the complex furin cleavage and processing observed in the native membrane-bound GP. Mice immunized with ZEBOVGP-Fc developed T-cell immunity against ZEBOV GP and neutralizing antibodies against replication-competent VSV-G deleted recombinant VSV containing ZEBOV GP. The ZEBOVGP-Fc vaccinated mice were protected against challenge with a lethal dose of ZEBOV. These results show that vaccination with the ZEBOVGP-Fc fusion protein alone without the need of a viral vector or assembly into virus-like particles is sufficient to induce protective immunity against ZEBOV in mice. Our data suggested that Filovirus GP Fc fusion proteins could be developed as a simple, safe, efficacious, and cost effective vaccine against Filovirus infection for human use.


Subject(s)
Ebola Vaccines/immunology , Hemorrhagic Fever, Ebola/prevention & control , Immunoglobulin Fc Fragments/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , CD8-Positive T-Lymphocytes/immunology , CHO Cells , Cricetinae , Cricetulus , Ebolavirus/immunology , HEK293 Cells , Hemorrhagic Fever, Ebola/immunology , Humans , Immunity, Humoral , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Processing, Post-Translational , Recombinant Fusion Proteins/immunology , Vaccines, Subunit/immunology , Viral Plaque Assay
7.
J Clin Invest ; 121(3): 1111-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21339644

ABSTRACT

During infection with the hepatitis A virus (HAV), most patients develop mild or asymptomatic disease. However, a small number of patients develop serious, life-threatening hepatitis. We investigated this variability in disease severity by examining 30 Argentinean patients with HAV-induced acute liver failure in a case-control, cross-sectional, observational study. We found that HAV-induced severe liver disease was associated with a 6-amino-acid insertion in TIM1/HAVCR1 (157insMTTTVP), the gene encoding the HAV receptor. This polymorphism was previously shown to be associated with protection against asthma and allergic diseases and with HIV progression. In binding assays, the TIM-1 protein containing the 157insMTTTVP insertion polymorphism bound HAV more efficiently. When expressed by human natural killer T (NKT) cells, this long form resulted in greater NKT cell cytolytic activity against HAV-infected liver cells, compared with the shorter TIM-1 protein without the polymorphism. To our knowledge, the 157insMTTTVP polymorphism in TIM1 is the first genetic susceptibility factor shown to predispose to HAV-induced acute liver failure. Furthermore, these results suggest that HAV infection has driven the natural selection of shorter forms of the TIM-1 protein, which binds HAV less efficiently, thereby protecting against severe HAV-induced disease, but which may predispose toward inflammation associated with asthma and allergy.


Subject(s)
Hepatitis A virus/metabolism , Hepatitis A/immunology , Liver Diseases/virology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Polymorphism, Genetic , Receptors, Virus/genetics , Receptors, Virus/physiology , Argentina , Case-Control Studies , Child , Child, Preschool , Cross-Sectional Studies , Female , Genetic Predisposition to Disease , Hepatitis A Virus Cellular Receptor 1 , Humans , Infant , Killer Cells, Natural/virology , Male , Risk
8.
J Virol ; 84(16): 8342-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20534860

ABSTRACT

Hepatitis A virus (HAV), an atypical member of the Picornaviridae, grows poorly in cell culture. To define determinants of HAV growth, we introduced a blasticidin (Bsd) resistance gene into the virus genome and selected variants that grew at high concentrations of Bsd. The mutants grew fast and had increased rates of RNA replication and translation but did not produce significantly higher virus yields. Nucleotide sequence analysis and reverse genetic studies revealed that a T6069G change resulting in a F42L amino acid substitution in the viral polymerase (3D(pol)) was required for growth at high Bsd concentrations whereas a silent C7027T mutation enhanced the growth rate. Here, we identified a novel determinant(s) in 3D(pol) that controls the kinetics of HAV growth.


Subject(s)
Hepatitis A virus/growth & development , Hepatitis A virus/genetics , Mutation, Missense , Viral Proteins/genetics , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , DNA Mutational Analysis , Macaca mulatta , Models, Molecular , Molecular Sequence Data , Point Mutation , Protein Structure, Tertiary , Sequence Alignment , Viral Plaque Assay
9.
J Immunol ; 184(4): 1918-30, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20083673

ABSTRACT

T cell/transmembrane, Ig, and mucin (TIM) proteins, identified using a congenic mouse model of asthma, critically regulate innate and adaptive immunity. TIM-1 and TIM-4 are receptors for phosphatidylserine (PtdSer), exposed on the surfaces of apoptotic cells. Herein, we show with structural and biological studies that TIM-3 is also a receptor for PtdSer that binds in a pocket on the N-terminal IgV domain in coordination with a calcium ion. The TIM-3/PtdSer structure is similar to that of TIM-4/PtdSer, reflecting a conserved PtdSer binding mode by TIM family members. Fibroblastic cells expressing mouse or human TIM-3 bound and phagocytosed apoptotic cells, with the BALB/c allelic variant of mouse TIM-3 showing a higher capacity than the congenic C.D2 Es-Hba-allelic variant. These functional differences were due to structural differences in the BC loop of the IgV domain of the TIM-3 polymorphic variants. In contrast to fibroblastic cells, T or B cells expressing TIM-3 formed conjugates with but failed to engulf apoptotic cells. Together these findings indicate that TIM-3-expressing cells can respond to apoptotic cells, but the consequence of TIM-3 engagement of PtdSer depends on the polymorphic variants of and type of cell expressing TIM-3. These findings establish a new paradigm for TIM proteins as PtdSer receptors and unify the function of the TIM gene family, which has been associated with asthma and autoimmunity and shown to modulate peripheral tolerance.


Subject(s)
Alleles , Apoptosis/immunology , Genetic Variation/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mucin-3/genetics , Phagocytosis/immunology , Phosphatidylserines/metabolism , Amino Acid Sequence , Animals , Apoptosis/genetics , Cell Line , Cell Line, Tumor , Hepatitis A Virus Cellular Receptor 2 , Humans , Membrane Proteins/physiology , Mice , Mice, Congenic , Mice, Inbred BALB C , Molecular Sequence Data , Mucin-3/metabolism , Mucin-3/physiology , Multigene Family/immunology , NIH 3T3 Cells , Phagocytosis/genetics
10.
Virol J ; 6: 204, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19922643

ABSTRACT

BACKGROUND: Hepatitis A virus (HAV), an atypical Picornaviridae that causes acute hepatitis in humans, grows poorly in cell culture and in general does not cause cytopathic effect. Foreign sequences have been inserted into different parts of the HAV genome. However, the packaging size limit of HAV has not been determined. The purpose of the present study is to investigate the maximum size of additional sequences that the HAV genome can tolerate without loosing infectivity. RESULTS: In vitro T7 polymerase transcripts of HAV constructs containing a 456-nt fragment coding for a blasticidin (Bsd) resistance gene, a 1,098-nt fragment coding for the same gene fused to GFP (GFP-Bsd), or a 1,032-nt fragment containing a hygromycin (Hyg) resistance gene cloned into the 2A-2B junction of the HAV genome were transfected into fetal Rhesus monkey kidney (FRhK4) cells. After antibiotic selection, cells transfected with the HAV construct containing the resistance gene for Bsd but not the GFP-Bsd or Hyg survived and formed colonies. To determine whether this size limitation was due to the position of the insertion, a 606 bp fragment coding for the Encephalomyocarditis virus (EMCV) internal ribosome entry site (IRES) sequence was cloned into the 5' nontranslated (NTR) region of HAV. The resulting HAV-IRES retained the EMCV IRES insertion for 1-2 passages. HAV constructs containing both the EMCV IRES at the 5' NTR and the Bsd-resistance gene at the 2A-2B junction could not be rescued in the presence of Bsd but, in the absence of antibiotic, the rescued viruses contained deletions in both inserted sequences. CONCLUSION: HAV constructs containing insertions of approximately 500-600 nt but not 1,000 nt produced viable viruses, which indicated that the HAV particles can successfully package approximately 600 nt of additional sequences and maintain infectivity.


Subject(s)
Hepatitis A virus/physiology , Mutagenesis, Insertional , RNA, Viral/genetics , Virus Assembly , Animals , Cell Line , Genetic Engineering , Hepatitis A virus/genetics , Macaca mulatta
11.
Virol J ; 6: 175, 2009 Oct 27.
Article in English | MEDLINE | ID: mdl-19860892

ABSTRACT

BACKGROUND: Hepatitis A virus (HAV), an atypical Picornaviridae that causes acute hepatitis in humans, usurps the HAV cellular receptor 1 (HAVCR1) to infect cells. HAVCR1 is a class 1 integral membrane glycoprotein that contains two extracellular domains: a virus-binding immunoglobulin-like (IgV) domain and a mucin-like domain that extends the IgV from the cell membrane. Soluble forms of HAVCR1 bind, alter, and neutralize cell culture-adapted HAV, which is attenuated for humans. However, the requirements of the HAV-HAVCR1 interaction have not been fully characterized, and it has not been determined whether HAVCR1 also serves as a receptor for wild-type (wt) HAV. Here, we used HAV soluble receptor neutralization and alteration assays to study the requirements of the HAV-HAVCR1 interaction and to determine whether HAVCR1 is also a receptor for wt HAV. RESULTS: Treatment of HAV with a soluble form of HAVCR1 that contained the IgV and two-thirds of the mucin domain fused to the Fc fragment of human IgG1 (D1 muc-Fc), altered particles at 37 degrees C but left a residual level of unaltered particles at 4 degrees C. The kinetics of neutralization of HAV by D1 muc-Fc was faster at 37 degrees C than at 4 degrees C. Alteration of HAV particles by D1 muc-Fc required Ca, which could not be replaced by Li, Na, Mg, Mn, or Zn. Neutralization of HAV by D1 muc-Fc occurred at pH 5 to 8 but was more efficient at pH 6 to 7. D1 muc-Fc neutralized wt HAV as determined by a cell culture system that allows the growth of wt HAV. CONCLUSION: The interaction of HAV with soluble forms of HAVCR1 shares the temperature, Ca, and pH requirements for infectivity in cell culture and therefore mimics the cell entry process of HAV. Since soluble forms of HAVCR1 also neutralized wt HAV, this receptor may play a significant role in pathogenesis of HAV.


Subject(s)
Hepatitis A virus/physiology , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , Virus Attachment , Animals , Cations/metabolism , Cell Line , Chlorocebus aethiops , Cricetinae , Cricetulus , Hepatitis A Virus Cellular Receptor 1 , Humans , Hydrogen-Ion Concentration , Kinetics , Membrane Glycoproteins/genetics , Metals/metabolism , Neutralization Tests , Protein Binding , Receptors, Virus/genetics , Temperature
12.
Virol J ; 5: 155, 2008 Dec 18.
Article in English | MEDLINE | ID: mdl-19094229

ABSTRACT

BACKGROUND: Hepatitis A virus (HAV), the causative agent of acute hepatitis in humans, is an atypical Picornaviridae that grows poorly in cell culture. HAV titrations are laborious and time-consuming because the virus in general does not cause cytopathic effect and is detected by immunochemical or molecular probes. Simple HAV titration assays could be developed using currently available viral construct containing selectable markers. RESULTS: We developed an antibiotic resistance titration assay (ARTA) based on the infection of human hepatoma cells with a wild type HAV construct containing a blasticidin (Bsd) resistance gene. Human hepatoma cells infected with the HAV-Bsd construct survived selection with 2 microg/ml of blasticidin whereas uninfected cells died within a few days. At 8 days postinfection, the color of the pH indicator phenol red in cell culture media correlated with the presence of HAV-Bsd-infected blasticidin-resistant cells: an orange-to-yellow color indicated the presence of growing cells whereas a pink-to-purple color indicated that the cells were dead. HAV-Bsd titers were determined by an endpoint dilution assay based on the color of the cell culture medium scoring orange-to-yellow wells as positive and pink-to-purple wells as negative for HAV. As a proof-of-concept, we used the ARTA to evaluate the HAV neutralization potency of two commercially available human immune globulin (IG) preparations and a WHO International Standard for anti-HAV. The three IG preparations contained comparable levels of anti-HAV antibodies that neutralized approximately 1.5 log of HAV-Bsd. Similar neutralization results were obtained in the absence of blasticidin by an endpoint dilution ELISA at 2 weeks postinfection. CONCLUSION: The ARTA is a simple and rapid method to determine HAV titers without using HAV-specific probes. We determined the HAV neutralization potency of human IG preparations in 8 days by ARTA compared to the 14 days required by the endpoint dilution ELISA. The ARTA reduced the labour, time, and cost of HAV titrations making it suitable for high throughput screening of sera and antivirals, determination of anti-HAV antibodies in human immune globulin preparations, and research applications that involve the routine evaluation of HAV titers.


Subject(s)
Antibodies, Viral/analysis , Drug Resistance, Microbial , Hepatitis A virus/isolation & purification , Hepatitis A/virology , Immunoglobulins/analysis , Virology/methods , Antibodies, Viral/immunology , Cell Line, Tumor , Hepatitis A/diagnosis , Hepatitis A/immunology , Hepatitis A virus/genetics , Hepatitis A virus/immunology , Humans , Immunoglobulins/immunology , Neutralization Tests/methods , Virus Cultivation/methods
13.
Immunity ; 27(6): 927-40, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18082433

ABSTRACT

The T cell immunoglobulin mucin (TIM) proteins regulate T cell activation and tolerance. Here we showed that TIM-4 is expressed on human and mouse macrophages and dendritic cells, and both TIM-4 and TIM-1 specifically bound phosphatidylserine (PS) on the surface of apoptotic cells but not any other phospholipid tested. TIM-4(+) peritoneal macrophages, TIM-1(+) kidney cells, and TIM-4- or TIM-1-transfected cells efficiently phagocytosed apoptotic cells, and phagocytosis could be blocked by TIM-4 or TIM-1 monoclonal antibodies. Mutations in the unique cavity of TIM-4 eliminated PS binding and phagocytosis. TIM-4 mAbs that blocked PS binding and phagocytosis mapped to epitopes in this binding cavity. These results show that TIM-4 and TIM-1 are immunologically restricted members of the group of receptors whose recognition of PS is critical for the efficient clearance of apoptotic cells and prevention of autoimmunity.


Subject(s)
Apoptosis , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Phagocytosis , Phosphatidylserines/metabolism , Receptors, Virus/metabolism , Animals , Dendritic Cells/metabolism , Hepatitis A Virus Cellular Receptor 1 , Humans , Lymphocyte Activation , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NIH 3T3 Cells , Phosphatidylserines/chemistry , T-Lymphocytes/immunology
14.
Immunity ; 27(6): 941-51, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18083575

ABSTRACT

The T cell immunoglobulin and mucin domain (TIM) proteins are important regulators of T cell responses. Crystal structures of the murine TIM-4 identified a metal-ion-dependent ligand binding site (MILIBS) in the immunoglobulin (Ig) domain of the TIM family. The characteristic CC' loop of the TIM domain and the hydrophobic FG loop shaped a narrow cavity where acidic compounds penetrate and coordinate to a metal ion bound to conserved residues in the TIM proteins. The structure of phosphatidylserine bound to the Ig domain showed that the hydrophilic head penetrates into the MILIBS and coordinates with the metal ion, whereas the aromatic residues on the tip of the FG loop interacted with the fatty acid chains and could insert into the lipid bilayer. Our results also revealed an important role of the MILIBS in the trafficking of TIM-1 to the cell surface.


Subject(s)
Membrane Proteins/chemistry , Metals/chemistry , Phosphatidylserines/metabolism , Amino Acid Sequence , Binding Sites , Cell Line , Crystallization , Humans , Immunoglobulin Variable Region/chemistry , Ligands , Membrane Proteins/metabolism , Molecular Sequence Data , Protein Transport
15.
Biochem Biophys Res Commun ; 356(4): 1017-23, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17407765

ABSTRACT

Hepatitis B virus (HBV), hepatitis C virus (HCV), and human immunodeficiency virus type-1 (HIV-1) are transfusion-transmitted human pathogens that have a major impact on blood safety and public health worldwide. We developed a microarray multiplex assay for the simultaneous detection and discrimination of these three viruses. The microarray consists of 16 oligonucleotide probes, immobilized on a silylated glass slide. Amplicons from multiplex PCR were labeled with Cy-5 and hybridized to the microarray. The assay detected 1 International Unit (IU), 10 IU, 20 IU of HBV, HCV, and HIV-1, respectively, in a single multiplex reaction. The assay also detected and discriminated the presence of two or three of these viruses in a single sample. Our data represent a proof-of-concept for the possible use of highly sensitive multiplex microarray assay to screen and confirm the presence of these viruses in blood donors and patients.


Subject(s)
DNA, Viral/blood , HIV-1/isolation & purification , Hepacivirus/isolation & purification , Hepatitis B virus/isolation & purification , Oligonucleotide Array Sequence Analysis/methods , Discriminant Analysis , HIV-1/genetics , Hepacivirus/genetics , Hepatitis B virus/genetics , Humans , Reproducibility of Results , Sensitivity and Specificity
16.
Immunity ; 26(3): 299-310, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17363299

ABSTRACT

The T cell immunoglobulin mucin (TIM) receptors are involved in the regulation of immune responses, autoimmunity, and allergy. Structures of the N-terminal ligand binding domain of the murine mTIM-1 and mTIM-2 receptors revealed an immunoglobulin (Ig) fold, with four Cys residues bridging a distinctive CC' loop to the GFC beta-sheet. The structures showed two ligand-recognition modes in the TIM family. The mTIM-1 structure identified a homophilic TIM-TIM adhesion interaction, whereas the mTIM-2 domain formed a dimer that prevented homophilic binding. Biochemical, mutational, and cell adhesion analyses confirmed the divergent ligand-binding modes revealed by the structures. Structural features characteristic of mTIM-1 appear conserved in human TIM-1, which also mediated homophilic interactions. The extracellular mucin domain enhanced binding through the Ig domain, modulating TIM receptor functions. These results explain the divergent immune functions described for the murine receptors and the role of TIM-1 as a cell adhesion receptor in renal regeneration and cancer.


Subject(s)
Membrane Proteins/chemistry , Amino Acid Sequence , Animals , Crystallography, X-Ray , Dimerization , Hepatitis A Virus Cellular Receptor 1 , Humans , Immunity , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Receptors, Immunologic/chemistry
17.
J Virol ; 81(7): 3437-46, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17229699

ABSTRACT

The hepatitis A virus cellular receptor 1 (HAVCR1/TIM1), a member of the T-cell immunoglobulin mucin (TIM) family, is an important atopy susceptibility gene in humans. The exact natural function of HAVCR1/TIM1 and the inverse association between HAV infection and prevention of atopy are not well understood. To identify natural ligands of human HAVCR1/TIM1, we used an expression cloning strategy based on the binding of dog cells transfected with a human lymph node cDNA library to a HAVCR1/TIM1 Fc fusion protein. The transfected cells that bound to the human HAVCR1/TIM1 Fc contained cDNA of human immunoglobulin alpha 1 heavy (Igalpha1) and lambda light (Iglambda) chain and secreted human IgA1lambda antibody that bound to the cell surface. Cotransfection of the isolated Igalpha1 and Iglambda cDNAs to naïve dog cells resulted in the secretion of IgA1lambda that bound to HAVCR1/TIM1 Fc but not to a poliovirus receptor Fc fusion protein in a capture enzyme-linked immunosorbent assay. The interaction of HAVCR1/TIM1 with IgA was inhibited by monoclonal antibodies (MAbs) against Igalpha1 and Iglambda, excess IgA1lambda, or anti-HAVCR1/TIM1 MAb. IgA did not inhibit HAV infection of African green monkey cells, suggesting that the IgA and the virus binding sites are in different epitopes on HAVCR1/TIM1. IgA enhanced significantly the neutralization of HAV by HAVCR1/TIM1 Fc. Our results indicate that IgA1lambda is a specific ligand of HAVCR1/TIM1 and that their association has a synergistic effect in virus-receptor interactions.


Subject(s)
Hepatitis A virus/immunology , Immunoglobulin A/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Receptors, Virus/immunology , Receptors, Virus/metabolism , Virus Internalization , Animals , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Chlorocebus aethiops , Cloning, Molecular , Cricetinae , Gene Expression , Hepatitis A Virus Cellular Receptor 1 , Humans , Ligands , Membrane Glycoproteins/genetics , Protein Binding , Receptors, Virus/genetics
18.
J Virol ; 80(3): 1352-60, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16415012

ABSTRACT

Human wild-type (wt) hepatitis A virus (HAV), the causative agent of acute hepatitis, barely grows in cell culture and in the process accumulates attenuating and cell culture-adapting mutations. This genetic instability of wt HAV in cell culture is a major roadblock to studying HAV pathogenesis and producing live vaccines that are not overly attenuated for humans. To develop a robust cell culture system capable of supporting the efficient growth of wt HAV, we transfected different cell lines with in vitro RNA transcripts of wt HAV containing the blasticidin resistance gene. Blasticidin-resistant colonies grew only in transfected Huh7 cells and produced infectious virus. HAV was genetically stable in Huh7 cells for at least nine serial passages and did not accumulate attenuating or cell culture-adapting mutations. Treatment with alpha interferon A/D cured the blasticidin-resistant Huh7 cells of the HAV infection. The cured cells, termed Huh7-A-I cells, did not contain virus or HAV antigens and were sensitive to blasticidin. Huh7-A-I cells were more permissive than parental cells for wt HAV infection, including a natural isolate from a human stool sample, and produced 10-fold-more infectious particles. This is the first report of a cell line that allows the genetically stable growth of human wt HAV. The viral vectors and cells described here should allow better insight into the pathogenesis of HAV and the development of attenuated vaccines. The cell lines susceptible to wt HAV growth may also be used to detect and isolate infectious virus from patient and environmental samples.


Subject(s)
Hepatitis A virus/physiology , Animals , Base Sequence , CHO Cells , Cell Line , Chlorocebus aethiops , Cricetinae , DNA, Viral/genetics , Genetic Vectors , HeLa Cells , Hepatitis A virus/genetics , Hepatitis A virus/growth & development , Hepatitis A virus/pathogenicity , Humans , Mice , Molecular Sequence Data , Transfection , Vero Cells , Virulence , Virus Cultivation/methods , Virus Replication
19.
J Virol ; 79(5): 2950-5, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15709014

ABSTRACT

Hepatitis A virus (HAV) has been adapted to grow efficiently in primate and some nonprimate cell lines but not in cells of murine origin. To understand the inability of the virus to grow in mouse cells, we studied the replication of HAV in immortalized and nontransformed MMH-D3 mouse liver cells, which require growth factors and collagen to maintain their phenotype. HAV grew in MMH-D3 cells transfected with virion RNA but not in those infected with viral particles, indicating a cell entry block for HAV. However, MMH-D3 cells cultured under suboptimal conditions in the absence of growth factors acquired susceptibility to HAV infection. Serial passages of the virus in MMH-D3 cells under suboptimal growth conditions resulted in the selection of HAV variants that grew efficiently in MMH-D3 cells cultured under both optimal and suboptimal conditions. Nucleotide sequence analysis of the MMH-D3 cell-adapted HAV revealed that N1237D and D2132G substitutions were present in the capsid regions of six viral clones. These two mutations are most likely located on the surface of the virion and may play a role in the entry of HAV into the mouse liver cells. Our results demonstrate that mouse hepatocyte-like cells code for all factors required for the efficient growth of HAV in cell culture.


Subject(s)
Hepatitis A virus/growth & development , Animals , Base Sequence , Capsid Proteins/genetics , Cell Culture Techniques , Cell Line , DNA, Viral/genetics , Genetic Variation , Hepatitis A virus/genetics , Hepatitis A virus/pathogenicity , Hepatitis A virus/physiology , Hepatocytes/virology , Mice , Mutation , RNA, Viral/genetics , Transfection , Virus Replication
20.
Kidney Int ; 65(5): 1761-73, 2004 May.
Article in English | MEDLINE | ID: mdl-15086915

ABSTRACT

BACKGROUND: The molecular mechanisms underlying tumorigenesis and progression of clear cell renal cell carcinoma (ccRCC) are not well understood. We aimed to identify new molecular markers to provide insight into these processes. METHODS: This work reports on the identification of human hepatitis A virus cellular receptor 1 (hHAVcr-1) as a differentially expressed gene in ccRCC using RNA-based arbitrarily primed polymerase chain reaction (RAP-PCR). Results were further confirmed by Northern and Western blot assays. Carcinoma 769-P and normal HK-2 cells derived from proximal tubule epithelial cells, grown under different culture conditions, were used to understand the putative role of hHAVcr-1 in renal malignancy. hHAVcr-1 stable transfected clones and dipeptidyl peptidase IV (DPPIV) assays allowed assessing its involvement in cell differentiation. RESULTS: The hHAVcr-1 is overexpressed in eight out of 13 ccRCC and its expression neglected in benign oncocytomas. In culture, hhavcr-1 is dramatically overexpressed in normal and tumor cell lines that, having acquired the fully differentiated phenotype, are induced to de-differentiate by means of phorbol ester phorbol 12-myristate-13-acetate (PMA) treatment. Similarly, differentiation prevention by addition of PMA to confluent cells also increases hhavcr-1 expression. hHAVcr-1 stable transfected 769-P cells proved that hhavcr-1 itself blocks differentiation. Since hhavcr-1 is expressed at higher levels in tumor cells, we used an African green monkey cell model to show that immunotoxins directed against the monkey homologue of hhavcr-1 could kill kidney cells. CONCLUSION: Our results showed that hHAVcr-1 blocks differentiation of proximal tubule epithelial cells and that it could be used as a target for therapy of kidney carcinomas.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/virology , Kidney Neoplasms/genetics , Kidney Neoplasms/virology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Receptors, Virus/genetics , Receptors, Virus/physiology , Adult , Aged , Aged, 80 and over , Animals , Base Sequence , Carcinoma, Renal Cell/pathology , Cell Differentiation , Cell Division , Cell Line, Tumor , Chlorocebus aethiops , Chromosome Mapping , Chromosomes, Human, Pair 5/genetics , DNA, Neoplasm/genetics , Female , Gene Expression , Hepatitis A Virus Cellular Receptor 1 , Hepatitis A virus/pathogenicity , Humans , In Situ Hybridization, Fluorescence , Kidney Neoplasms/pathology , Male , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...