Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Hepatology ; 62(5): 1511-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26175263

ABSTRACT

UNLABELLED: Autoimmune hepatitis (AIH) is known as a T cell-mediated disease. However, AIH patients refractory to conventional treatment have been successfully treated with anti-CD20-mediated B-cell depletion. The aim of this project was to understand the immunological changes underlying the AIH remission caused by B-cell depletion in an experimental model of AIH. C57BL/6 AIH mice, xenoimmunized with DNA coding for human liver antigens, were treated with a single dose of depleting mouse anti-CD20 antibody at the peak of liver inflammation. Liver inflammation, alanine aminotransferase levels, chemokine (C-X-C) ligand 10 expression, and circulating B-cell, autoantibody, and total immunoglobulin G levels were monitored following depletion. T-cell and B-cell phenotype and function were characterized. Administration of a single dose of anti-CD20 resulted in a drastic reduction of liver inflammation accompanied by a significant reduction of alanine aminotransferase levels and of proinflammatory chemokine (C-X-C) ligand 10 expression. The treatment did not result in significant changes in total immunoglobulin G levels or autoantibodies. There were significantly more naive and less antigen-experienced CD4+ and CD8+ T cells, and T-cell proliferation was significantly reduced following anti-CD20 treatment. B cells served as antigen-presenting cells to CD4+ T cells. Anti-CD20 treatment also led to a profound reduction of T follicular helper cells. CONCLUSION: B cells play an active role in the pathogenesis of AIH in antigen presentation processes and the modulation of T-cell functions and influence the T follicular helper-cell population; this active role of B cells could explain the success of B-cell depletion for remission of AIH despite its classification as a T cell-mediated autoimmune liver disease.


Subject(s)
Antigen Presentation , B-Lymphocytes/physiology , Hepatitis, Autoimmune/immunology , Lymphocyte Depletion , T-Lymphocytes/immunology , Animals , Antigens, CD20/immunology , Autoantigens/immunology , Cytokines/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL
2.
Liver Int ; 35(1): 275-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24517723

ABSTRACT

BACKGROUND: Some patients with autoimmune hepatitis (AIH), despite appropriate treatment, progress towards cirrhosis and liver failure, requiring transplantation. New biological agents targeting immune cell subtypes have been developed, with better specificity and longer-lasting effects than conventional wide-spectrum immunosuppressive drugs. AIMS: The goal of this study was to evaluate the effectiveness of low dose of αCD3 targeting therapy in a model of type 2 AIH. METHODS: This experimental model is based on xenoimmunization of C57BL/6 mice with DNA coding for human liver autoantigens. Mice with AIH were treated with five daily injections of low dose of αCD3 monoclonal antibody, before disease onset (5.5 months post-xenoimmunization) or during AIH (7 months post-xenoimmunization). Along with serum aminotransferases, autoantibody levels and end-point liver histology, spleen and liver-infiltrating lymphocytes were phenotyped by flow cytometry and immune response measured by lymphoproliferative assays. RESULTS: Before onset of AIH, treatment prevented the development of liver inflammation and tissue injury. During active AIH, low dose of αCD3 antibody therapy resulted in a resorption of liver inflammatory infiltrates, normalization of serum aminotransferas levels, reduced autoantibody titres, increased regulatory T cells and lowered proliferation of autoreactive liver lymphocytes. CONCLUSIONS: We report that low dose αCD3 antibody administration is an effective treatment for AIH in an experimental model of type 2 AIH. These data suggest that αCD3 antibody therapy could be tested in clinical trials as a rescue therapy for patients with uncontrolled AIH.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD3 Complex/immunology , Hepatitis, Autoimmune/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Autoantigens/administration & dosage , Flow Cytometry , Mice , Mice, Inbred C57BL , Remission Induction/methods , Transaminases/blood
3.
Brain ; 134(Pt 12): 3560-77, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22058139

ABSTRACT

Clonally expanded CD8(+) T lymphocytes are present in multiple sclerosis lesions, as well as in the cerebrospinal fluid of patients with multiple sclerosis. In experimental autoimmune encephalomyelitis, CD8(+) T lymphocytes are found in spinal cord and brainstem lesions. However, the exact phenotype of central nervous system-infiltrating CD8(+) T lymphocytes and the mechanism by which these cells cross the blood-brain barrier remain largely unknown. Using cerebrospinal fluid from patients with multiple sclerosis, spinal cord from experimental autoimmune encephalomyelitis and coronavirus-induced encephalitis, we demonstrate that central nervous system-infiltrating CD8(+) T lymphocytes are mostly of the effector memory phenotype (CD62L(-) CCR7(-) granzymeB(hi)). We further show that purified human effector memory CD8(+) T lymphocytes transmigrate more readily across blood-brain barrier-endothelial cells than non-effector memory CD8(+) T lymphocytes, and that blood-brain barrier endothelium promotes the selective recruitment of effector memory CD8(+) T lymphocytes. Furthermore, we provide evidence for the recruitment of interferon-γ- and interleukin-17-secreting CD8(+) T lymphocytes by human and mouse blood-brain barrier endothelium. Finally, we show that in vitro migration of CD8(+) T lymphocytes across blood-brain barrier-endothelial cells is dependent on α4 integrin, but independent of intercellular adhesion molecule-1/leucocyte function-associated antigen-1, activated leucocyte cell adhesion molecule/CD6 and the chemokine monocyte chemotactic protein-1/CCL2. We also demonstrate that in vivo neutralization of very late antigen-4 restricts central nervous system infiltration of CD8(+) T lymphocytes in active immunization and adoptive transfer experimental autoimmune encephalomyelitis, and in coronavirus-induced encephalitis. Our study thus demonstrates an active role of the blood-brain barrier in the recruitment of effector memory CD8(+) T lymphocytes to the CNS compartment and defines α4 integrin as a major contributor of CD8(+) T lymphocyte entry into the brain.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Coronavirus Infections/immunology , Encephalitis, Viral/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4/metabolism , Multiple Sclerosis/immunology , Adult , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/immunology , Coronavirus Infections/metabolism , Encephalitis, Viral/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Immunologic Memory , Mice , Middle Aged , Multiple Sclerosis/metabolism
4.
J Neurovirol ; 16(4): 279-93, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20642316

ABSTRACT

The etiology of most neurodegenerative diseases of the central nervous system remains unknown and likely involves a combination of genetic susceptibility and environmental triggering factors. Given that exposure to numerous infectious pathogens occurs during childhood, and that some viral infections can lead to neurodegeneration and demyelination, it is conceivable that some viruses may act as triggering factors in neuropathogenesis. We have previously shown that the prototype OC43 strain of the common cold-associated human respiratory coronavirus has the capacity to infect human neuronal and glial cells and does persist in human brains. Moreover, it has neuroinvasive properties in susceptible BALB/c mice, where it leads to a chronic encephalitis with accompanying disabilities. Here, we show that mutations in the viral spike glycoprotein, reproducibly acquired during viral persistence in human neural cell cultures, led to a drastically modified virus-induced neuropathology in BALB/c mice, characterized by flaccid paralysis and demyelination. Even though infection by both mutated and wild-type viruses led to neuroinflammation, the modified neuropathogenesis induced by the mutated virus was associated with increased viral spread and significantly more CD4+ and CD8+ T-lymphocyte infiltration into the central nervous system, as well as significantly increased levels of the proinflammatory cytokine interleukin (IL)-6 and the chemokine CCL2 (monocyte chemoattractant protein [MCP]-1). Moreover, recombinant virus harboring the S glycoprotein mutations retained its neurotropism, productively infecting neurons. Therefore, interaction of a human respiratory coronavirus with the central nervous system may modulate virus and host factors resulting in a modified neuropathogenesis in genetically susceptible individuals.


Subject(s)
Coronavirus Infections/genetics , Coronavirus OC43, Human/pathogenicity , Demyelinating Diseases/virology , Encephalitis, Viral/genetics , Membrane Glycoproteins/genetics , Mutation , Paraplegia/virology , Viral Envelope Proteins/genetics , Animals , Coronavirus Infections/complications , Coronavirus Infections/pathology , Demyelinating Diseases/pathology , Encephalitis, Viral/complications , Encephalitis, Viral/pathology , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Paraplegia/pathology , Spike Glycoprotein, Coronavirus
5.
J Vis Exp ; (14)2008 Apr 28.
Article in English | MEDLINE | ID: mdl-19066576

ABSTRACT

Determination of infectious viral titers is a basic and essential experimental approach for virologists. Classical plaque assays cannot be used for viruses that do not cause significant cytopathic effects, which is the case for prototype strains 229E and OC43 of human coronavirus (HCoV).Therefore, an alternative indirect immunoperoxidase assay (IPA) was developed for the detection and titration of these viruses and is described herein. Susceptible cells are inoculated with serial logarithmic dilutions of virus-containing samples in a 96-well plate format. After viral growth,viral detection by IPA yields the infectious virus titer, expressed as 'Tissue Culture Infectious Dose 50 percent' (TCID50). This represents the dilution of a virus-containing sample at which half of a series of laboratory wells contain infectious replicating virus. This technique provides are liable method for the titration of HCoV-229E and HCoV-OC43 in biological samples such as cells, tissues and fluids [corrected].


Subject(s)
Coronavirus 229E, Human/chemistry , Coronavirus OC43, Human/chemistry , Immunoenzyme Techniques/methods , Viral Proteins/analysis , Cells, Cultured , Common Cold/pathology , Common Cold/virology , Coronavirus 229E, Human/isolation & purification , Coronavirus Infections/pathology , Coronavirus Infections/virology , Coronavirus OC43, Human/isolation & purification , Humans , Reproducibility of Results , Titrimetry/methods , Viral Proteins/biosynthesis
6.
Methods Mol Biol ; 454: 93-102, 2008.
Article in English | MEDLINE | ID: mdl-19057861

ABSTRACT

Calculation of infectious viral titers represents a basic and essential experimental approach for virologists. Classical plaque assays cannot be used for viruses that do not cause significant cytopathic effects, which is the case for strains 229E and OC43 of human coronavirus (HCoV). An alternative indirect immunoperoxidase assay (IPA) is herein described for the detection and titration of these viruses. Susceptible cells are inoculated with serial logarithmic dilutions of samples in a 96-well plate. After viral growth, viral detection by IPA yields the infectious virus titer, expressed as "Tissue Culture Infectious Dose" (TCID50). This represents the dilution of a virus-containing sample at which half of a series of laboratory wells contain replicating virus. This technique is a reliable method for the titration of HCoV in biological samples (cells, tissues, or fluids).


Subject(s)
Coronavirus 229E, Human/isolation & purification , Coronavirus OC43, Human/isolation & purification , Immunoenzyme Techniques/methods , Coronavirus 229E, Human/immunology , Coronavirus OC43, Human/immunology , Humans , Reproducibility of Results
7.
Hepatology ; 42(3): 675-82, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16037945

ABSTRACT

Anti-liver-kidney microsome type 1 (LKM1) autoantibodies directed against the cytochrome P450 2D6 (CYP2D6) are considered specific markers of type 2 autoimmune hepatitis, but are also found in 5% of sera from patients chronically infected by hepatitis C virus (HCV). Molecular mimicry between HCV proteins and CYP2D6 has been proposed to explain the emergence of these autoantibodies. Anti-LKM1 autoantibodies from hepatitis C-infected patients were affinity-purified against immobilized CYP2D6 protein and used to screen a phage display library. CYP2D6 conformational epitopes were identified using phage display analysis and the identification of statistically significant pairs (SSPs). Cross-reactivity between CYP2D6 and HCV protein candidates was tested by immunoprecipitation. Nineteen different clones were isolated, and their sequencing resulted in the mapping of a conformational epitope to the region of amino acids 254-288 of CYP2D6. Candidate HCV proteins for molecular mimicry included: core, E2, NS3 and NS5a. Affinity-purified autoantibodies from HCV+/LKM1+ patients immunoprecipitated either NS3, NS5a, or both, and these reactivities were specifically inhibited by immobilized CYP2D6. In conclusion, HCV+/LKM1+ sera recognize a specific conformational epitope on CYP2D6 between amino acids 254 to 288, the region that contains the major linear epitope in type 2 autoimmune hepatitis patients. Cross-reactivity due to molecular mimicry at the B-cell level was shown between the CYP2D6 and the HCV NS3 and NS5a proteins and could explain the presence of anti-LKM1 in patients chronically infected with HCV. Further investigation of the role played by this molecular mimicry in HCV-infected patients may lead to more specific strategies for diagnosis and treatment.


Subject(s)
Autoantibodies/blood , Hepatitis C, Chronic/immunology , Molecular Mimicry/immunology , Amino Acid Sequence , Autoantibodies/immunology , Cytochrome P-450 CYP2D6/chemistry , Cytochrome P-450 CYP2D6/genetics , Female , Hepacivirus/genetics , Humans , Male , Models, Molecular , Protein Conformation , Viral Proteins/genetics
8.
Autoimmunity ; 37(3): 217-22, 2004 May.
Article in English | MEDLINE | ID: mdl-15497455

ABSTRACT

Hepatitis C infection is associated with autoimmune disorders, such as the production of autoantibodies. Anti-LKM1 and anti-LC1, immunomarkers of type 2 autoimmune hepatitis, have been previously associated with a HCV infection. Anti-Soluble-Liver-Antigen autoantibodies (SLA) are specifically associated with type 1 and type 2 autoimmune hepatitis and more closely related to patients who relapse after steroid therapy. The recent molecular cloning of the soluble liver antigen provides the opportunity to develop more specific tests for the detection of antibodies against it. The aim of this work is to characterize anti-soluble-liver autoantibodies in sera from patients chronically infected by HCV. A recombinant cDNA from activated Jurkat cells coding for the full length tRNP(Ser)Sec/SLA antigen was obtained. ELISA, Western Blot and immunoprecipitation tests were developed and used to search for linear and conformational epitopes recognized by anti-SLA antibodies in sera from patients chronically infected by HCV. Anti-soluble liver antigen antibodies were found in sera from 10.4% of HCV-infected patients. The prevalence was significantly increased to 27% when anti-LKM1 was also present. Most anti-SLA reactivity was directed against conformational epitopes on the antigen. The means titers by ELISA were lower than those obtained in type 2 AIH. The result of autoantibody isotyping showed a subclass restriction to IgG1 and also IgG4. This study shows the presence of anti-SLA antibodies in approximately 10% of HCV infected patients. The prevalence of SLA autoantibodies in HCV infected patients increases when LKM1 autoantibodies are also present. The relationship between the prevalence of this characteristic autoimmune hepatitis autoantibody and the implication of an autoimmune phenomenon in the liver injury of patients chronically infected by HCV needs further investigation.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Hepatitis C/immunology , Enzyme-Linked Immunosorbent Assay , Hepatitis, Autoimmune/immunology , Humans
9.
J Autoimmun ; 22(2): 159-66, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14987745

ABSTRACT

UNLABELLED: Various autoantibodies have been reported in patients chronically infected by hepatitis C virus. 2% to 10% of theses patients have anti-liver-kidney microsome type 1 (anti-LKM1) autoantibodies. In type 2 autoimmune hepatitis, anti-LKM1 autoantibodies are frequently associated with anti-liver-cytosol type 1 (anti-LC1) autoantibodies. AIMS: To determine the prevalence of anti-LC1 autoantibodies in a hepatitis C-positive population and characterize their reactivity. METHODS: 146 patients suffering from liver diseases, of which 99 were chronically infected by hepatitis C virus, were tested by Western blotting and immunoprecipitation to detect and characterize anti-LC1 autoantibodies. RESULTS: 12% of this hepatitis C population had anti-LC1 autoantibodies. LC1 positivity by Western blotting was 30% of LC1+ sera. Epitopes were found throughout the protein but linear epitopes were situated in the 395-541 amino acid region of formiminotransferase cyclodeaminase. Three putative conformational epitopes were identified by phage display. CONCLUSIONS: Anti-LC1 autoantibodies are as prevalent as anti-LKM1 autoantibodies in patients infected with hepatitis C virus and their production is not dependent of anti-LKM1 autoantibodies formation. Autoantibody reactivity against the anti-LC1 antigen is different in hepatitis C than in type 2 autoimmune hepatitis. Anti-LC1 autoantibodies can now be regarded as a serological marker of autoimmunity in chronic hepatitis C infection.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Adult , Ammonia-Lyases/genetics , Ammonia-Lyases/immunology , Autoantibodies/blood , Autoantigens/genetics , Child, Preschool , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/immunology , Epitope Mapping , Hepatitis C, Chronic/blood , Hepatitis, Autoimmune/blood , Hepatitis, Autoimmune/immunology , Humans , Recombinant Proteins/genetics , Recombinant Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...