Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 15(3): 488-97, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23474736

ABSTRACT

Recent commercial approval of cancer vaccine, demonstrating statistically significant improvement in overall survival of prostate cancer patients has spurred renewed interest in active immunotherapies; specifically, strategies that lead to enhanced biological activity and robust efficacy for dendritic cell vaccines. A simple, widely used approach to generating multivalent cancer vaccines is to load tumor whole cell lysates into dendritic cells (DCs). Current DC vaccine manufacturing processes require co-incubation of tumor lysate antigens with immature DCs and their subsequent maturation. We compared electroloading of tumor cell lysates directly into mature DCs with the traditional method of lysate co-incubation with immature DCs. Electroloaded mature DCs were more potent in vitro, as judged by their ability to elicit significantly (p < 0.05) greater expansion of peptide antigen-specific CD8(+) T cells, than either lysate-electroloaded immature DCs or lysate-co-incubated immature DCs, both of which must be subsequently matured. Expanded CD8(+) T cells were functional as judged by their ability to produce IFN-γ upon antigen-specific re-stimulation. The electroloading technology used herein is an automated, scalable, functionally closed cGMP-compliant manufacturing technology supported by a Master File at CBER, FDA and represents an opportunity for translation of enhanced potency DC vaccines at clinical/commercial scale.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/metabolism , Electroporation/methods , Immunotherapy, Adoptive/methods , Melanoma/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation , Antigens, Neoplasm/immunology , CD8 Antigens/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Feasibility Studies , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Melanoma/therapy
2.
PLoS One ; 7(3): e32367, 2012.
Article in English | MEDLINE | ID: mdl-22412866

ABSTRACT

Tularemia or vaccination with the live vaccine strain (LVS) of Francisella tularensis confers long-lived cell-mediated immunity. We hypothesized that this immunity depends on polyfunctional memory T cells, i.e., CD4(+) and/or CD8(+) T cells with the capability to simultaneously express several functional markers. Multiparametric flow cytometry, measurement of secreted cytokines, and analysis of lymphocyte proliferation were used to characterize in vitro recall responses of peripheral blood mononuclear cells (PBMC) to killed F. tularensis antigens from the LVS or Schu S4 strains. PBMC responses were compared between individuals who had contracted tularemia, had been vaccinated, or had not been exposed to F. tularensis (naïve). Significant differences were detected between either of the immune donor groups and naïve individuals for secreted levels of IL-5, IL-6, IL-10, IL-12, IL-13, IFN-γ, MCP-1, and MIP-1ß. Expression of IFN-γ, MIP-1ß, and CD107a by CD4(+)CD45RO(+) or CD8(+)CD45RO(+) T cells correlated to antigen concentrations. In particular, IFN-γ and MIP-1ß strongly discriminated between immune and naïve individuals. Only one cytokine, IL-6, discriminated between the two groups of immune individuals. Notably, IL-2- or TNF-α-secretion was low. Our results identify functional signatures of T cells that may serve as correlates of immunity and protection against F. tularensis.


Subject(s)
Francisella tularensis/immunology , T-Lymphocytes/immunology , Tularemia/immunology , Adult , Age Factors , Aged , Antigens, Bacterial/immunology , Cytokines/metabolism , Epitopes/immunology , Female , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Male , Middle Aged , Sex Factors , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Tularemia/metabolism , Tularemia/prevention & control , Young Adult
3.
Int J Med Microbiol ; 301(7): 591-601, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21873113

ABSTRACT

Francisella tularensis is pathogenic for many mammalian species including humans, causing a spectrum of diseases called tularemia. The highly virulent Type A strains have associated mortality rates of up to 60% if inhaled. An attenuated live vaccine strain (LVS) is the only vaccine to show efficacy in humans, but suffers several barriers to licensure, including the absence of a correlate of protection. An immunoproteomics approach was used to survey the repertoire of antibodies in sera from individuals who had contracted tularemia during two outbreaks and individuals from two geographical areas who had been vaccinated with NDBR Lot 11 or Lot 17 LVS. These data showed a large overlap in the antibodies generated in response to tularemia infection or LVS vaccination. A total of seven proteins were observed to be reactive with 60% or more sera from vaccinees and convalescents. A further four proteins were recognised by 30-60% of the sera screened. These proteins have the potential to serve as markers of vaccination or candidates for subunit vaccines.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Vaccines/immunology , Francisella tularensis/immunology , Proteome/analysis , Tularemia/immunology , Humans , Vaccines, Attenuated/immunology
4.
Eur J Immunol ; 41(4): 974-80, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21442618

ABSTRACT

The efficacy of many vaccines against intracellular bacteria depends on the generation of cell-mediated immunity, but studies to determine the duration of immunity are usually confounded by re-exposure. The causative agent of tularemia, Francisella tularensis, is rare in most areas and, therefore, tularemia vaccination is an interesting model for studies of the longevity of vaccine-induced cell-mediated immunity. Here, lymphocyte proliferation and cytokine production in response to F. tularensis were assayed in two groups of 16 individuals, vaccinated 1-3 or 27-34 years previously. As compared to naïve individuals, vaccinees of both groups showed higher proliferative responses and, out of 17 cytokines assayed, higher levels of MIP-1ß, IFN-γ, IL-10, and IL-5 in response to recall stimulation. The responses were very similar in the two groups of vaccinees. A statistical model was developed to predict the immune status of the individuals and by use of two parameters, proliferative responses and levels of IFN-γ, 91.1% of the individuals were correctly classified. Using flow cytometry analysis, we demonstrated that during recall stimulation, expression of IFN-γ by CD4(+) CCR7(+) , CD4(+) CD62L(+) , CD8(+) CCR7(+) , and CD8(+) CD62L(+) cells significantly increased in samples from vaccinated donors. In conclusion, cell-mediated immunity was found to persist three decades after tularemia vaccination without evidence of decline.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , Adult , Antigens, Bacterial/immunology , Cell Proliferation , Cells, Cultured , Cytokines/biosynthesis , Cytokines/immunology , Female , Humans , Lymphocytes/cytology , Lymphocytes/immunology , Male , Vaccines, Attenuated/immunology
5.
Hepatology ; 45(1): 31-41, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187411

ABSTRACT

UNLABELLED: Oval cells are hepatocytic precursors that proliferate in late-stage cirrhosis and that give rise to a subset of human hepatocellular carcinomas. Although liver regeneration typically occurs through replication of existing hepatocytes, oval cells proliferate only when hepatocyte proliferation is inhibited. Transforming growth factor-beta (TGF-beta) is a key inhibitory cytokine for hepatocytes, both in vitro and in vivo. Because TGF-beta levels are elevated in chronic liver injury when oval cells arise, we hypothesized that oval cells may be less responsive to the growth inhibitory effects of this cytokine. To examine TGF-beta signaling in vivo in oval cells, we analyzed livers of rats fed a choline-deficient, ethionine-supplemented (CDE) diet for phospho-Smad2. Phospho-Smad2 was detected in more than 80% of hepatocytes, but staining was substantially reduced in oval cells. Ki67 staining, in contrast, was significantly more common in oval cells than hepatocytes. To understand the inverse relationship between TGF-beta signaling and proliferation in oval cells and hepatocytes, we examined TGF-beta signaling in vitro. TGF-beta caused marked growth inhibition in primary hepatocytes and the AML12 hepatocyte cell line. Two oval cell lines, LE/2 and LE/6, were less responsive. The greater sensitivity of the hepatocytes to TGF-beta-induced growth inhibition may result from the absence of Smad6 in these cells. CONCLUSION: Our results indicate that oval cells, both in vivo and in vitro, are less sensitive to TGF-beta-induced growth inhibition than hepatocytes. These findings further suggest an underlying mechanism for the proliferation of oval cells in an environment inhibitory to hepatocytic proliferation.


Subject(s)
Cell Proliferation , Hepatocytes/cytology , Stem Cells/metabolism , Transforming Growth Factor beta/physiology , Animals , Apoptosis , Cells, Cultured , Cyclins/genetics , Cyclins/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation , Hepatocytes/metabolism , Mice , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Smad Proteins/genetics , Smad Proteins/metabolism , Stem Cells/cytology
6.
J Immunol ; 177(12): 8301-5, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17142725

ABSTRACT

TCR engagement leads to the up-regulation of genetic programs that can both activate and inhibit T cell function. The early growth receptor (Egr) proteins Egr-2 and Egr-3 have recently been identified as TCR-induced negative regulators of T cell function. NAB2 (NGFI-A-binding protein 2) is both a coactivator and a corepressor of Egr-mediated transcription and has been implicated in regulating Schwann cell myelination. In this report we demonstrate that NAB2 is induced by TCR engagement and that its expression is enhanced by the presence of costimulation. The overexpression of NAB2 enhanced IL-2 production while small interfering RNA to NAB2 markedly inhibited IL-2 expression. Mechanistically, we demonstrate that NAB2 enhances IL-2 transcription by acting as a coactivator for Egr-1. Indeed, chromatin immunoprecipitation analysis reveals that NAB2 is recruited to the Egr-1 binding site of the IL-2 promoter. Taken together, our findings identify NAB2 as a novel coactivator of T cell function.


Subject(s)
Interleukin-2/genetics , Neoplasm Proteins/immunology , Receptors, Antigen, T-Cell/physiology , Repressor Proteins/immunology , Animals , Early Growth Response Protein 1/metabolism , Gene Expression Regulation/immunology , Interleukin-2/biosynthesis , Mice , Mice, Inbred Strains , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/physiology , Transcription, Genetic
7.
Arch Immunol Ther Exp (Warsz) ; 54(1): 1-13, 2006.
Article in English | MEDLINE | ID: mdl-16642252

ABSTRACT

The first line of treatment for many human autoimmune diseases involves the use of anti-inflammatory or immunosuppressive drugs such as prednisone or other steroids that not only suppress the underlying autoimmune disease, but lead to global suppression of the immune system. The sequelae of this approach include increased risk of infection, carcinogenesis, and osteoporosis. Moreover, such broad spectrum immunosuppression tends to have transient therapeutic benefit, as in many cases the disease becomes refractory to these drugs. There is a clear need for more specific means to restore immune tolerance to the specific autoantigens implicated in disease pathology. This review provides an overview of some of these newer, more specific therapeutic approaches to restoring immune tolerance to autoantigens, with an emphasis on those approaches that have been or will soon be tested in controlled clinical trials. Covered here are peptide- or protein-based therapeutics, oral tolerance, and cellular and gene therapy approaches to restoring antigen-specific immune tolerance.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/therapy , Immune Tolerance , Immunosuppressive Agents/therapeutic use , Adoptive Transfer , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , B-Lymphocytes/immunology , Dendritic Cells/immunology , Genetic Therapy , Humans , Immunosuppressive Agents/adverse effects , T-Lymphocytes, Regulatory/immunology
8.
Mol Ther ; 13(1): 42-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16219491

ABSTRACT

Hematopoietic stem cell (HSC) transplantation is a potential therapy that can offer multiple sclerosis patients a radical, potentially curative treatment. Using experimental autoimmune encephalomyelitis (EAE) as a model, we previously reported that retrovirally transduced B cells expressing myelin basic protein (MBP), MBP Ac1-11, or myelin oligodendrocyte glycoprotein p35-55 induced tolerance and reduced symptoms. Here, we extend our tolerance approach using bone marrow (BM) cells expressing full-length phospholipid protein (PLP) in a model for relapsing, remitting EAE. Using GFP expression as a marker, we found that up to 50% of cells were positive for transgene expression in peripheral blood after 900 rad irradiation and transduced BM transplantation, and expression was stable in hematopoietic lineages for over 10 weeks. Upon challenge, T cell proliferation in response to PLP p139-151 was reduced and EAE was completely abolished in a pretreatment protocol. In addition, protection from EAE could be achieved with PLP-transduced BM cells given on day 12 after immunization, a potential therapeutic protocol. Finally, the protective effect of PLP-expressing BM could also be observed using a nonmyeloablative protocol, albeit with lower efficacy. Our results suggest that HSC may be useful to achieve long-lasting tolerance to protect mice from EAE and possibly to promote CNS repair in ongoing EAE.


Subject(s)
Bone Marrow Transplantation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Therapy , Immune Tolerance , Myelin Proteolipid Protein/metabolism , Peptide Fragments/metabolism , Animals , Bone Marrow Cells/metabolism , CD4 Antigens/immunology , Cell Proliferation , Combined Modality Therapy , Encephalomyelitis, Autoimmune, Experimental/radiotherapy , Female , Genetic Vectors , Green Fluorescent Proteins/genetics , Humans , Immunization , L-Selectin/immunology , Mice , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/immunology , Peptide Fragments/genetics , Peptide Fragments/immunology , Receptors, Interleukin-2/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
9.
J Immunol ; 174(5): 2481-4, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15728451

ABSTRACT

Cell cycle re-entry of quiescent T cells is dependent upon cyclin-dependent kinase 2. Inhibition of cyclin-dependent kinase 2 by p27(Kip1) is believed to be the principal constraint on S-phase entry in T cells. We report that deficiency for p27(Kip1) has a more pronounced effect on the expansion of murine naive CD8(+) T cells and that this disparity is due to a reduced requirement for CD28-mediated costimulation in CD8(+) but not CD4(+) T cells lacking p27(Kip1). These data highlight a previously unappreciated difference in the way CD28 signaling is coupled to the core cell cycle machinery in these two T cell subsets.


Subject(s)
CD28 Antigens/physiology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle Proteins/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Lymphocyte Activation/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Animals , CD28 Antigens/genetics , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle Proteins/biosynthesis , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Female , Kinetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Suppressor Proteins/biosynthesis
10.
N Engl J Med ; 351(6): 552-9, 2004 Aug 05.
Article in English | MEDLINE | ID: mdl-15295048

ABSTRACT

BACKGROUND: The receptors for transforming growth factor beta (TGF-beta) and their signaling intermediates make up an important tumor-suppressor pathway. The role of one of these intermediates--Smad3--in the pathogenesis of lymphoid neoplasia is unknown. METHODS: We measured Smad3 messenger RNA (mRNA) and protein in leukemia cells obtained at diagnosis from 19 children with acute leukemia, including 10 with T-cell acute lymphoblastic leukemia (ALL), 7 with pre-B-cell ALL, and 2 with acute nonlymphoblastic leukemia (ANLL). All nine exons of the SMAD3 gene (MADH3) were sequenced. Mice in which one or both alleles of Smad3 were inactivated were used to evaluate the role of Smad3 in the response of normal T cells to TGF-beta and in the susceptibility to spontaneous leukemogenesis in mice in which both alleles of the tumor suppressor p27Kip1 were deleted. RESULTS: Smad3 protein was absent in T-cell ALL but present in pre-B-cell ALL and ANLL. No mutations were found in the MADH3 gene in T-cell ALL, and Smad3 mRNA was present in T-cell ALL and normal T cells at similar levels. In mice, the loss of one allele for Smad3 impairs the inhibitory effect of TGF-beta on the proliferation of normal T cells and works in tandem with the homozygous inactivation of p27Kip1 to promote T-cell leukemogenesis. CONCLUSIONS: Loss of Smad3 protein is a specific feature of pediatric T-cell ALL. A reduction in Smad3 expression and the loss of p27Kip1 work synergistically to promote T-cell leukemogenesis in mice.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , Leukemia, T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/metabolism , T-Lymphocytes/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics , Adult , Animals , Cell Cycle Proteins/metabolism , Child , Cyclin-Dependent Kinase Inhibitor p27 , DNA-Binding Proteins/genetics , Exons , Gene Deletion , Gene Expression , Humans , Interleukin-2/biosynthesis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Mice , Mice, Knockout , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Transforming Growth Factor beta/metabolism , Sequence Analysis, DNA , Signal Transduction , Smad3 Protein , Trans-Activators/genetics , Tumor Suppressor Proteins/metabolism
11.
J Immunol ; 173(5): 3093-102, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15322169

ABSTRACT

Induction of G(1) arrest by TGF-beta correlates with the regulation of p21(Cip1) and p27(Kip1), members of the Cip/Kip family of cyclin-dependent kinase inhibitors (cki). However, no definitive evidence exists that these proteins play a causal role in TGF-beta(1)-induced growth arrest in lymphocytes. In this report we show the suppression of cell cycle progression by TGF-beta is diminished in T cells from mice deficient for both p21(Cip1) and p27(Kip1) (double-knockout (DKO)) only when activated under conditions of optimal costimulation. Although there is an IL-2-dependent enhanced proliferation of CD8(+) T cells from DKO mice, TGF-beta is able to maximally suppress the proliferation of DKO T cells when activated under conditions of low costimulatory strength. We also show that the induction of p15(Ink4b) in T cells stimulated in the presence of TGF-beta is not essential, as TGF-beta also efficiently suppressed proliferation of T cells from p15(Ink4b-/-) mice. Finally, although these cki are dispensable for the suppression of T cell proliferation by TGF-beta, we now describe a Smad3-dependent down-regulation of cdk4, suggesting a potential mechanism underlying to resistance of Smad3(-/-) T cells to the induction of growth arrest by TGF-beta. In summary, the growth suppressive effects of TGF-beta in naive T cells are a function of the strength of costimulation, and alterations in the expression of cki modify the sensitivity to TGF-beta by lowering thresholds for a maximal mitogenic response.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclins/metabolism , G1 Phase/physiology , Interleukin-2/metabolism , Proto-Oncogene Proteins , T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle Proteins/immunology , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/immunology , Cyclin-Dependent Kinases/metabolism , Cyclins/immunology , Down-Regulation , G1 Phase/immunology , Immunologic Memory/immunology , Immunologic Memory/physiology , Interleukin-2/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Knockout , T-Lymphocytes/immunology , Transforming Growth Factor beta/immunology , Tumor Suppressor Proteins/immunology , Up-Regulation
12.
J Immunol Methods ; 266(1-2): 7-18, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12133618

ABSTRACT

Administration of transforming growth factor-beta (TGF-beta) has been found to be of therapeutic benefit in various mouse disease models and has potential clinical usefulness. However, the ability to track the distribution of exogenously administered, recombinant forms of these proteins has been restricted by cross-reactivity with endogenous TGF-beta and related TGF-beta isoforms. We describe novel FLAG- and hemagglutinin (HA)-tagged versions of mature TGF-beta1 that retain full biological activity as demonstrated by their ability to inhibit the growth of Mv1Lu epithelial cells, and to induce phosphorylation of the TGF-beta signaling intermediate, smad 2. Intracellular FLAG- and HA-TGF-beta1 can be detected in transfected cells by confocal immunofluorescence microscopy. We also describe sandwich ELISAs designed to specifically detect epitope-tagged TGF-beta and demonstrate the utility of these tagged ligands as probes for TGF-beta receptor expression by flow cytometry. The design of these fully functional epitope-tagged TGF-beta proteins should facilitate studies such as the evaluation of in vivo peptide pharmacodynamics and trafficking of TGF-beta ligand-receptor complexes.


Subject(s)
Transforming Growth Factor beta/analysis , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cell Line , Enzyme-Linked Immunosorbent Assay/methods , Epitopes/analysis , Flow Cytometry/methods , Hemagglutinins/genetics , Hemagglutinins/immunology , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Molecular Sequence Data , Oligopeptides , Peptides/genetics , Peptides/immunology , Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Transforming Growth Factor beta/analysis , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins , Transfection , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...