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










Database
Language
Publication year range
1.
Mol Ther Methods Clin Dev ; 2: 15029, 2015.
Article in English | MEDLINE | ID: mdl-26445723

ABSTRACT

Adeno-associated virus (AAV) has become one of the most promising vectors in gene transfer in the last 10 years with successful translation to clinical trials in humans and even market approval for a first gene therapy product in Europe. Administration to humans, however, revealed that adaptive immune responses against the vector capsid can present an obstacle to sustained transgene expression due to the activation and expansion of capsid-specific T cells. The limited number of peripheral blood mononuclear cells (PBMCs) obtained from samples within clinical trials allows for little more than monitoring of T-cell responses. We were able to identify immunodominant major histocompatibility complex (MHC) class I epitopes for common human leukocyte antigen (HLA) types by using spleens isolated from subjects undergoing splenectomy for non-malignant indications as a source of large numbers of lymphocytes and restimulating them with single AAV capsid peptides in vitro. Further experiments confirmed that these epitopes are naturally processed and functionally relevant. The design of more effective and less immunogenic AAV vectors, and precise immune monitoring of vector-infused subjects, are facilitated by these findings.

2.
Mol Ther ; 21(9): 1727-37, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23857231

ABSTRACT

Immune responses directed against viral capsid proteins constitute a main safety concern in the use of adeno-associated virus (AAV) as gene transfer vectors in humans. Pharmacological immunosuppression has been proposed as a solution to the problem; however, the approach suffers from several potential limitations. Using MHC class II epitopes initially identified within human IgG, named Tregitopes, we showed that it is possible to modulate CD8+ T cell responses to several viral antigens in vitro. We showed that incubation of peripheral blood mononuclear cells with these epitopes triggers proliferation of CD4+CD25+FoxP3+ T cells that suppress killing of target cells loaded with MHC class I antigens in an antigen-specific fashion, through a mechanism that seems to require cell-to-cell contact. Expression of a construct encoding for the AAV capsid structural protein fused to Tregitopes resulted in reduction of CD8+ T cell reactivity against the AAV capsid following immunization with an adenoviral vector expressing capsid. This was accompanied by an increase in frequency of CD4+CD25+FoxP3+ T cells in spleens and lower levels of inflammatory infiltrates in injected tissues. This proof-of-concept study demonstrates modulation of CD8+ T cell reactivity to an antigen using regulatory T cell epitopes is possible.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/immunology , Dependovirus/immunology , Epitopes, T-Lymphocyte/immunology , Genetic Vectors , Immunoglobulin G/immunology , Animals , Antigens, Viral/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Capsid/immunology , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cells, Cultured , Dependovirus/genetics , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Genetic Therapy , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunoglobulin G/genetics , Male , Mice , Mice, Inbred C57BL , Spleen/immunology , T-Lymphocytes, Regulatory/immunology
3.
Allergy Asthma Proc ; 31(6): 507-10, 2010.
Article in English | MEDLINE | ID: mdl-20943012

ABSTRACT

Egg allergy is a contraindication for influenza vaccination because of the presence of trace egg protein. Although recent reports suggest that egg-allergic individuals can be safely immunized with seasonal influenza vaccine, the safety of H1N1 influenza vaccine in this context has not previously been reported. This study was designed to investigate how to safely immunize high-risk populations during the H1N1 influenza pandemic for whom the vaccine would be contraindicated. Vaccination of egg-allergic children was conducted at a large, multispecialty practice. Pediatricians raised awareness of vaccination and referred egg-allergic individuals to allergists. Allergists performed skin testing to H1N1 vaccine and, if negative, performed graded vaccine desensitizations in the office. Mass immunization of egg-allergic individuals was achieved within a 3-week period after receipt of vaccine. Sixty-three children (average age, 3.7 years) were evaluated and skin tested to the H1N1 influenza vaccine. All had a clinical history of egg allergy and recent positive skin-prick to egg. All had negative skin-prick test to the H1N1 vaccine and received graded challenge with three divided doses. All 62 children were successfully immunized without allergic reactions. Vaccination of egg-allergic individuals during a pandemic can be safely and efficiently accomplished with the coordination of multidisciplinary resources. Although this was conducted at a multispecialty practice, we believe that this invaluable public health service can be done outside of this context. Allergists, with the help of local pediatricians and urgent care centers or hospitals, can effectively mobilize to help protect the most vulnerable populations during pandemics.


Subject(s)
Egg Hypersensitivity/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Adolescent , Child , Child, Preschool , Contraindications , Desensitization, Immunologic , Egg Hypersensitivity/complications , Egg Hypersensitivity/diagnosis , Egg Hypersensitivity/therapy , Female , Humans , Infant , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/complications , Influenza, Human/diagnosis , Influenza, Human/prevention & control , Interdisciplinary Communication , Male , Mass Vaccination , Pandemics , Skin Tests
4.
Mol Ther ; 18(1): 135-42, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19904235

ABSTRACT

Adeno-associated viral (AAV) vectors are an extensively studied and highly used vector platform for gene therapy applications. We hypothesize that in the first clinical trial using AAV to treat hemophilia B, AAV capsid proteins were presented on the surface of transduced hepatocytes, resulting in clearance by antigen-specific CD8+ T cells and consequent loss of therapeutic transgene expression. It has been previously shown that proteasome inhibitors can have a dramatic effect on AAV transduction in vitro and in vivo. Here, we describe using the US Food and Drug Administration-approved proteasome inhibitor, bortezomib, to decrease capsid antigen presentation on hepatocytes in vitro, whereas at the same time, enhancing gene expression in vivo. Using an AAV capsid-specific T-cell reporter (TCR) line to analyze the effect of proteasome inhibitors on antigen presentation, we demonstrate capsid antigen presentation at low multiplicities of infection (MOIs), and inhibition of antigen presentation at pharmacologic levels of bortezomib. We also demonstrate that bortezomib can enhance Factor IX (FIX) expression from an AAV2 vector in mice, although the same effect was not observed for AAV8 vectors. A pharmacological agent that can enhance AAV transduction, decrease T-cell activation/proliferation, and decrease capsid antigen presentation would be a promising solution to obstacles to successful AAV-mediated, liver-directed gene transfer in humans.


Subject(s)
Antigen Presentation/drug effects , Boronic Acids/therapeutic use , Capsid Proteins/immunology , Dependovirus/genetics , Genetic Vectors/genetics , Protease Inhibitors/therapeutic use , Pyrazines/therapeutic use , Transduction, Genetic/methods , Animals , Bortezomib , CD8-Positive T-Lymphocytes/immunology , Cell Line , Dependovirus/immunology , Genes, MHC Class I/genetics , Hemophilia B/therapy , Humans , Mice
5.
J Clin Invest ; 119(6): 1688-95, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19436115

ABSTRACT

Adeno-associated virus (AAV) vectors are effective gene delivery vehicles mediating long-lasting transgene expression. Data from a clinical trial of AAV2-mediated hepatic transfer of the Factor IX gene (F9) into hemophilia B subjects suggests that CTL responses against AAV capsid can eliminate transduced hepatocytes and prevent long-term F9 expression. However, the capacity of hepatocytes to present AAV capsid-derived antigens has not been formally demonstrated, nor whether transduction by AAV sensitizes hepatocytes for CTL-mediated destruction. To investigate the fate of capsids after transduction, we engineered a soluble TCR for the detection of capsid-derived peptide:MHC I (pMHC) complexes. TCR multimers exhibited antigen and HLA specificity and possessed high binding affinity for cognate pMHC complexes. With this reagent, capsid pMHC complexes were detectable by confocal microscopy following AAV-mediated transduction of human hepatocytes. Although antigen presentation was modest, it was sufficient to flag transduced cells for CTL-mediated lysis in an in vitro killing assay. Destruction of hepatocytes was inhibited by soluble TCR, demonstrating a possible application for this reagent in blocking undesirable CTL responses. Together, these studies provide a mechanism for the loss of transgene expression and transient elevations in aminotransferases following AAV-mediated hepatic gene transfer in humans and a potential therapeutic intervention to abrogate these limitations imposed by the host T cell response.


Subject(s)
Antigen Presentation/immunology , Capsid Proteins/immunology , Cytotoxicity, Immunologic/immunology , Dependovirus/genetics , Genetic Vectors/genetics , Hepatocytes/immunology , Hepatocytes/metabolism , Capsid Proteins/metabolism , Cell Line , Hepatocytes/cytology , Histocompatibility Antigens/immunology , Humans , Protein Multimerization , Receptors, Antigen, T-Cell/immunology , Solubility , Substrate Specificity , T-Lymphocytes, Cytotoxic/immunology
6.
Ann Allergy Asthma Immunol ; 100(4): 392-5, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18450128

ABSTRACT

Increases in total serum IgE levels can be observed in many diverse conditions, from infection to atopy to primary immunodeficiency. The differentiation of atopy from immunodeficiency most often is made on a clinical basis, after taking findings from history, physical examination, and laboratory studies into consideration. However, total IgE level is neither a sensitive nor a specific diagnostic marker for any particular disease and, therefore, should not be relied on to establish a diagnosis of either atopy or primary immunodeficiency.


Subject(s)
Dermatitis, Atopic/diagnosis , Hypergammaglobulinemia/diagnosis , Immunoglobulin E/blood , Immunologic Deficiency Syndromes/diagnosis , Dermatitis, Atopic/immunology , Diagnosis, Differential , Humans , Hypergammaglobulinemia/immunology , Immunoglobulin E/immunology , Immunologic Deficiency Syndromes/immunology
7.
FASEB J ; 20(7): 896-905, 2006 May.
Article in English | MEDLINE | ID: mdl-16675847

ABSTRACT

Leukocyte trafficking to the central nervous system (CNS), regulated in part by chemokines, determines severity of the demyelinating diseases multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). To examine chemokine receptor CX3CR1 in EAE, we studied CX3CR1(GFP/GFP) mice, in which CX3CR1 targeting by insertion of Green Fluorescent Protein (GFP) allowed tracking of CX3CR1+ cells in CX3CR1(+/GFP) animals and cells destined to express CX3CR1 in CX3CR1(GFP/GFP) knockouts. NK cells were markedly reduced in the inflamed CNS of CX3CR1-deficient mice with EAE, whereas recruitment of T cells, NKT cells and monocyte/macrophages to the CNS during EAE did not require CX3CR1. Impaired recruitment of NK cells in CX3CR1(GFP/GFP) mice was associated with increased EAE-related mortality, nonremitting spastic paraplegia and hemorrhagic inflammatory lesions. The absence of CD1d did not affect the severity of EAE in CX3CR1(GFP/GFP) mice, arguing against a role for NKT cells. Accumulation of NK cells in livers of wild-type (WT) and CX3CR1(GFP/GFP) mice with cytomegalovirus hepatitis was equivalent, indicating that CX3CL1 mediated chemoattraction of NK cells was relatively specific for the CNS. These results are the first to define a chemokine that governs NK cell migration to the CNS, and the findings suggest novel therapeutic manipulation of CX3CR1+ NK cells.


Subject(s)
Central Nervous System/metabolism , Chemokines, CX3C/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Membrane Proteins/metabolism , Animals , Antigens, CD1/metabolism , Antigens, CD1d , Brain Stem/pathology , Central Nervous System/pathology , Chemokine CX3CL1 , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Hemorrhage/pathology , Killer Cells, Natural/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Paraparesis, Spastic/physiopathology , Spinal Cord/pathology
8.
J Immunol ; 169(10): 5827-37, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12421964

ABSTRACT

The kinetics of CD8 T cell IFN-gamma responses as they occur in situ are defined here during lymphocytic choriomeningitis virus (LCMV) infections, and a unique mechanism for the innate cytokines IFN-alphabeta and IL-18 in promoting these responses is defined. Infections of mice with Armstrong or WE strains of LCMV induced an unexpectedly early day 4 IFN-gamma response detectable in serum samples and spleen and liver homogenates. Production of IFN-gamma was MHC class I/CD8 dependent, but did not require IL-12, NK cells, TCR-gammadelta T cells, MHC class II, or CD4 T cells. Peak response required specific Ag recognition, as administration of antagonist peptide partially impaired day 4 IFN-gamma induction, and viral peptide stimulation enhanced CD8 T cell IFN-gamma expression in culture. The IFN-gamma response was associated with IL-18 and IFN-alphabeta expression. Furthermore, both factors augmented peptide-driven IFN-gamma production in culture, and mice lacking IL-18 or IFN-alphabeta functions had reduced day 4 IFN-gamma. Collectively, these results demonstrate that during viral infections, there is a dramatic in vivo CD8 T cell response preceding maximal expansion of these cells, and that the mechanism supporting this response is dependent on endogenous innate cytokines. Because stimulation by microbial products is linked to innate cytokine expression, the studies also suggest a pathway for precisely limiting T cell functions to times of need.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/physiology , Lymphocytic Choriomeningitis/immunology , Animals , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Egg Proteins/physiology , Immunity, Innate/genetics , Immunodominant Epitopes/physiology , Interferon-gamma/biosynthesis , Interleukin-12/physiology , Interleukin-18/physiology , Kinetics , Lymphocyte Activation/genetics , Lymphocytic Choriomeningitis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleoproteins/immunology , Nucleoproteins/physiology , Ovalbumin/physiology , Peptide Fragments/immunology , Peptide Fragments/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Time Factors
9.
Science ; 297(5589): 2063-6, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12242445

ABSTRACT

Interferons (IFNs) are essential for host defense. Although the antiviral effects of the type 1 IFNs IFN-alpha and IFN-beta (IFN-alpha/beta) have been established, their immunoregulatory functions, especially their ability to regulate IFN-gamma production, are poorly understood. Here we show that IFN-alpha/beta activate STAT4 directly (STAT, signal transducers and activators of transcription) and that this is required for IFN-gamma production during viral infections of mice, in concert with T cell receptor-derived signals. In contrast, STAT1 appears to negatively regulate IFN-alpha/beta induction of IFN-gamma. Thus, type 1 IFNs, in addition to interleukin-12, provide pathways for innate regulation of adaptive immunity, and their immunoregulatory functions are controlled by modulating the activity of individual STATs.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Interferon Type I/immunology , Interferon-gamma/biosynthesis , Lymphocytic choriomeningitis virus , Trans-Activators/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Female , Gene Expression Regulation , Interferon Type I/pharmacology , Interferon-gamma/genetics , Interleukin-12/physiology , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Promoter Regions, Genetic , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Recombinant Proteins , STAT1 Transcription Factor , STAT4 Transcription Factor , Signal Transduction , Th1 Cells/immunology , Th1 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...