Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
J Neuroimmunol ; 339: 577136, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31855721

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease affecting the neuromuscular junction. Approximately 9% of MG patients have autoantibodies targeting the muscle specific kinase (MuSK), and are challenging therapeutically, since they often present with more severe symptoms. A useful therapy is plasmapheresis, but it is highly non-specific. Antigen-specific immunoadsorption would only remove the pathogenic autoantibodies, minimizing the possible side effects and maximizing the benefit. We used rats with human MuSK-induced experimental autoimmune MG to perform antigen-specific immunoadsorptions, and found it very effective, resulting in a dramatic autoantibody titer decrease, while immunoadsorbed, but not mock-treated, animals showed an significant improvement of their clinical symptoms. Overall, the procedure was efficient, supporting its application for MG treatment.


Subject(s)
Autoantibodies/administration & dosage , Histocompatibility Antigens Class II/administration & dosage , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Plasmapheresis/methods , Receptor Protein-Tyrosine Kinases/administration & dosage , Receptors, Cholinergic/administration & dosage , Animals , Autoantibodies/immunology , Female , Histocompatibility Antigens Class II/immunology , Humans , Immunization/methods , Myasthenia Gravis, Autoimmune, Experimental/immunology , Rats , Rats, Inbred Lew , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Cholinergic/immunology , Treatment Outcome
2.
Transl Stroke Res ; 11(4): 831-836, 2020 08.
Article in English | MEDLINE | ID: mdl-31797249

ABSTRACT

Recognizing that the pathologic progression of stroke is closely associated with aberrant immune responses, in particular the activation of peripheral leukocytes, namely T cells, we hypothesized that finding a treatment designed to inhibit neuroantigen-specific T cells and block cytotoxic monocytes and macrophages may render therapeutic effects in stroke. We previously reported that subcutaneous administration of partial MHC class II constructs promote behavioral and histological effects in stroke mice by centrally promoting a protective M2 macrophage/microglia phenotype in the CNS and peripherally reversing stroke-associated splenic atrophy. Here, we employed a second species using adult Sprague-Dawley rats exposed to the middle cerebral artery occlusion stroke model and observed similar therapeutic effects with a mouse partial MHC class II construct called DRmQ, as evidenced by reductions in stroke-induced motor deficits, infarcts, and peri-infarct cell loss and neuroinflammation. More importantly, we offered further evidence of peripheral sequestration of inflammation at the level of the spleen, which was characterized by attenuation of stroke-induced spleen weight reduction and TNF-ɑ and IL-6 upregulation. Collectively, these results satisfy the Stroke Therapy Academic Industry Roundtable criteria of testing a novel therapeutic in a second species and support the use of partial MHC class II constructs as a stroke therapeutic designed to sequester both central and peripheral inflammation responses in an effort to retard, or even halt, the neuroinflammation that exacerbates the secondary cell death in stroke.


Subject(s)
Histocompatibility Antigens Class II/administration & dosage , Inflammation/prevention & control , Neuroprotective Agents/administration & dosage , Stroke/prevention & control , Animals , Encephalitis/prevention & control , Inflammation/complications , Inflammation/metabolism , Male , Rats, Sprague-Dawley , Stroke/complications , Stroke/pathology
3.
Cancer Res ; 78(12): 3280-3292, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29588348

ABSTRACT

Cancer immunotherapies utilize distinct mechanisms to harness the power of the immune system to eradicate cancer cells. Therapeutic vaccines, aimed at inducing active immune responses against an existing cancer, are highly dependent on the immunological microenvironment, where many immune cell types display high levels of plasticity and, depending on the context, promote very different immunologic outcomes. Among them, plasmacytoid dendritic cells (pDC), known to be highly immunogenic upon inflammation, are maintained in a tolerogenic state by the tumor microenvironment. Here, we report that intratumoral (i.t.) injection of established solid tumors with CpG oligonucleotides-B (CpG-B) inhibits tumor growth. Interestingly, control of tumor growth was independent of tumor-associated pDC, which remained refractory to CpG-B stimulation and whose depletion did not alter the efficacy of the treatment. Instead, tumor growth inhibition subsequent to i.t. CpG-B injection depended on the recruitment of neutrophils into the milieu, resulting in the activation of conventional dendritic cells, subsequent increased antitumor T-cell priming in draining lymph nodes, and enhanced effector T-cell infiltration in the tumor microenvironment. These results reinforce the concept that i.t. delivery of TLR9 agonists alters the tumor microenvironment by improving the antitumor activity of both innate and adaptive immune cells.Significance: Intratumoral delivery of CpG-B disrupts the tolerogenic tumor microenvironment and inhibits tumor growth. Cancer Res; 78(12); 3280-92. ©2018 AACR.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Immune Tolerance/drug effects , Neoplasms/therapy , Oligodeoxyribonucleotides/administration & dosage , Tumor Microenvironment/drug effects , Animals , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Communication/drug effects , Cell Communication/immunology , Cell Line, Tumor/transplantation , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/immunology , Humans , Immunotherapy/methods , Injections, Intralesional , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Neutrophils/drug effects , Neutrophils/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptor 9/agonists , Tumor Microenvironment/immunology
4.
Biol Pharm Bull ; 39(8): 1309-18, 2016 Aug 01.
Article in English | MEDLINE | ID: mdl-27251665

ABSTRACT

An adequate immune response to percutaneous vaccine application is generated by delivery of sufficient amounts of antigen to skin and by administration of toxin adjuvants or invasive skin abrasion that leads to an adjuvant effect. Microneedles penetrate the stratum corneum, the outermost layer of the skin, and enable direct delivery of vaccines from the surface into the skin, where immunocompetent dendritic cells are densely distributed. However, whether the application of microneedles to the skin activates antigen-presenting cells (APCs) has not been demonstrated. Here we aimed to demonstrate that microneedles may act as a potent physical adjuvant for successful transcutaneous immunization (TCI). We prepared samples of isolated epidermal and dermal cells and analyzed the expression of major histocompatibility complex (MHC) class II and costimulatory molecules on Langerhans or dermal dendritic cells in the prepared samples using flow cytometry. The expression of MHC class II and costimulatory molecules demonstrated an upward trend in APCs in the skin after the application of 500- and 300-µm microneedles. In addition, in the epidermal cells, application of microneedles induced more effective activation of Langerhans cells than did an invasive tape-stripping (positive control). In conclusion, the use of microneedles is likely to have a positive effect not only as an antigen delivery system but also as a physical technique inducing an adjuvant-like effect for TCI.


Subject(s)
Dendritic Cells/immunology , Langerhans Cells/immunology , Microinjections , Needles , Skin/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, CD/administration & dosage , Histocompatibility Antigens Class II/administration & dosage , Immunization , Injections, Subcutaneous , Male , Mice , Mice, Inbred C57BL , Skin/cytology
6.
Sci Rep ; 5: 11820, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26139288

ABSTRACT

The development of an efficacious Plasmodium falciparum malaria vaccine remains a top priority for global health. Vaccination with irradiated sporozoites is able to provide complete sterile protection through the action of CD8(+) T cells at the liver-stage of infection. However, this method is currently unsuitable for large-scale deployment and focus has instead turned to the development of sub-unit vaccines. Sub-unit vaccine efforts have traditionally focused on two well-known pre-erythrocytic antigens, CSP and TRAP, yet thousands of genes are expressed in the liver-stage. We sought to assess the ability of eight alternative P. falciparum pre-erythrocytic antigens to induce a high proportion of CD8(+) T cells. We show that all antigens, when expressed individually in the non-replicating viral vectors ChAd63 and MVA, are capable of inducing an immune response in mice. Furthermore, we also developed chimeric P. berghei parasites expressing the cognate P. falciparum antigen to enable assessment of efficacy in mice. Our preliminary results indicate that vectors encoding either PfLSA1 or PfLSAP2 are capable of inducing sterile protection dependent on the presence of CD8(+) T cells. This work has identified two promising P. falciparum liver-stage candidate antigens that will now undergo further testing in humans.


Subject(s)
Antigens, Protozoan/immunology , Histocompatibility Antigens Class II/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Animals , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors , Histocompatibility Antigens Class II/administration & dosage , Humans , Liver/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mice , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Vaccination
7.
Methods Mol Biol ; 1139: 321-36, 2014.
Article in English | MEDLINE | ID: mdl-24619690

ABSTRACT

A novel method for amplifying the activity of major histocompatibility complex (MHC) class II helper epitopes entails linking a 4-amino-acid moiety (LRMK) from the invariant chain (Ii) of MHC (referred to as Ii-Key) to the N-terminal end of the epitope peptide either directly or using a simple polymethylene spacer (-ava-). Ii-Key catalyzes binding of the linked epitope to the MHC class II molecule, thereby enhancing the overall potency of presentation. HER-2(776-790) (or AE36), which is derived from the intracellular domain of HER-2/neu, has been intensively used as an Ii-key/HER-2(776-790) (or AE37) fusion (hybrid) vaccine in clinical trials. This chapter describes procedures for the synthesis, reconstitution, sterility testing, and storage of both AE36 and AE37 for their use in clinical trials. Also provided is a detailed information about their in vivo administration and analysis of in-depth protocols for monitoring of immune activation upon vaccination with AE37.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Cancer Vaccines/immunology , Histocompatibility Antigens Class II/immunology , Receptor, ErbB-2/immunology , Vaccines, Subunit/immunology , Amino Acid Sequence , Antigens, Differentiation, B-Lymphocyte/administration & dosage , Antigens, Differentiation, B-Lymphocyte/chemistry , Antigens, Differentiation, B-Lymphocyte/genetics , Breast Neoplasms/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Cancer Vaccines/genetics , Cell Proliferation , Female , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Humans , Molecular Sequence Data , Ovarian Neoplasms/immunology , Protein Structure, Tertiary , Receptor, ErbB-2/administration & dosage , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics
8.
ACS Nano ; 7(4): 3036-44, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23540530

ABSTRACT

Here we present a biomimetic strategy toward nanoparticle design for controlled immune response through encapsulation of conserved internal influenza proteins on the interior of virus-like particles (VLPs) to direct CD8+ cytotoxic T cell protection. Programmed encapsulation and sequestration of the conserved nucleoprotein (NP) from influenza on the interior of a VLP, derived from the bacteriophage P22, results in a vaccine that provides multistrain protection against 100 times lethal doses of influenza in an NP specific CD8+ T cell-dependent manner. VLP assembly and encapsulation of the immunogenic NP cargo protein is the result of a genetically programmed self-assembly making this strategy amendable to the quick production of vaccines to rapidly emerging pathogens. Addition of adjuvants or targeting molecules were not required for eliciting the protective response.


Subject(s)
Histocompatibility Antigens Class II/administration & dosage , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Vaccines, Virus-Like Particle/administration & dosage , Animals , Biomimetic Materials/administration & dosage , Biomimetic Materials/chemical synthesis , Histocompatibility Antigens Class II/chemistry , Influenza A Virus, H1N1 Subtype , Mice , Treatment Outcome , Vaccines, Virus-Like Particle/chemistry , Vaccines, Virus-Like Particle/immunology
9.
J Immunol ; 187(9): 4492-500, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21949026

ABSTRACT

Accumulation of amyloid-ß peptide (Aß) is considered the triggering factor of pathogenic lesions in Alzheimer's disease (AD), and vaccines targeting Aß are promising therapeutic options. However, the occurrence of meningoencephalitides attributed to T cell responses in 6% of Aß-immunized patients underscores the need for a better understanding of T cell responses to Aß. We characterized the parameters controlling the magnitude of Aß-specific CD4(+) T cell responses in mice. T cell responsiveness to Aß1-42 was highly heterogeneous between mouse strains of different H-2 haplotypes, with SJL/J (H-2(s)) mice displaying a strong response, mainly specific for Aß10-24, and C57BL/6 (H-2(b)) mice displaying a weak response to Aß16-30. Surprisingly, C57BL/6 mice congenic for the H-2(s) haplotype (B6.H-2(S)), which display a "permissive" MHC class II allele for presentation of the immunodominant Aß10-24 epitope, showed a very weak CD4(+) T cell response to Aß, suggesting that MHC-independent genes downmodulate Aß-specific CD4(+) T cell responses in C57BL/6 background. Vaccine-induced CD4(+) T cell responses to Aß were significantly enhanced in both C57BL/6 and B6.H-2(S) mice upon depletion of regulatory T cells (Tregs), whereas Treg-depleted SJL/J mice displayed unaltered Aß-specific T cell responses. Finally, Treg depletion in C57BL/6 transgenic APPPS1 mice, a mouse model of AD, results in enhanced vaccine-induced CD4(+) T cell responses in AD compared with wild-type animals. We concluded that the magnitude of Aß-specific CD4(+) T cell responses is critically controlled in both physiological and pathological settings by MHC-independent genetic factors that determine the overall potency of Aß-specific Treg responses.


Subject(s)
Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/antagonists & inhibitors , CD4-Positive T-Lymphocytes/immunology , H-2 Antigens/genetics , Peptide Fragments/administration & dosage , Peptide Fragments/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Alleles , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Peptides/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/pathology , Dose-Response Relationship, Immunologic , Epitopes, T-Lymphocyte/immunology , H-2 Antigens/immunology , Histocompatibility Antigens Class II/administration & dosage , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Mice, Transgenic , Molecular Sequence Data , Peptide Fragments/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/pathology
10.
J Virol ; 85(13): 6442-52, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21490092

ABSTRACT

Major histocompatibility complex (MHC) molecules expressed on the surface of human immunodeficiency virus (HIV) are potential targets for neutralizing antibodies. Since MHC molecules are polymorphic, nonself MHC can also be immunogenic. We have used combinations of novel recombinant HLA class I and II and HIV/simian immunodeficiency virus (SIV) antigens, all linked to dextran, to investigate whether they can elicit protective immunity against heterologous simian/human immunodeficiency virus (SHIV) challenge in rhesus macaques. Three groups of animals were immunized with HLA (group 1, n = 8), trimeric YU2 HIV type 1 (HIV-1) gp140 and SIV p27 (HIV/SIV antigens; group 2, n = 8), or HLA plus HIV/SIV antigens (group 3, n = 8), all with Hsp70 and TiterMax Gold adjuvant. Another group (group 4, n = 6) received the same vaccine as group 3 without TiterMax Gold. Two of eight macaques in group 3 were completely protected against intravenous challenge with 18 50% animal infective doses (AID(50)) of SHIV-SF162P4/C grown in human cells expressing HLA class I and II lineages represented in the vaccine, while the remaining six macaques showed decreased viral loads compared to those in unimmunized animals. Complement-dependent neutralizing activity in serum and high levels of anti-HLA antibodies were elicited in groups 1 and 3, and both were inversely correlated with the plasma viral load at 2 weeks postchallenge. Antibody-mediated protection was strongly supported by the fact that transfer of pooled serum from the two challenged but uninfected animals protected two naïve animals against repeated low-dose challenge with the same SHIV stock. This study demonstrates that immunization with recombinant HLA in combination with HIV-1 antigens might be developed into an alternative strategy for a future AIDS vaccine.


Subject(s)
AIDS Vaccines/administration & dosage , Gene Products, gag/administration & dosage , HIV Infections/prevention & control , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class I/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , env Gene Products, Human Immunodeficiency Virus/administration & dosage , Animals , Female , Gene Products, gag/genetics , Gene Products, gag/immunology , HIV Infections/immunology , HIV-1/immunology , HIV-1/pathogenicity , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Immunization , Macaca mulatta/immunology , Molecular Sequence Data , Recombination, Genetic , SAIDS Vaccines/administration & dosage , Sequence Analysis, DNA , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Treatment Outcome , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology
11.
J Immunol ; 186(4): 2355-64, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21257961

ABSTRACT

Potent and broad cellular immune responses against the nonstructural (NS) proteins of hepatitis C virus (HCV) are associated with spontaneous viral clearance. In this study, we have improved the immunogenicity of an adenovirus (Ad)-based HCV vaccine by fusing NS3 from HCV (Strain J4; Genotype 1b) to the MHC class II chaperone protein invariant chain (Ii). We found that, after a single vaccination of C57BL/6 or BALB/c mice with Ad-IiNS3, the HCV NS3-specific CD8(+) T cell responses were significantly enhanced, accelerated, and prolonged compared with the vaccine encoding NS3 alone. The AdIiNS3 vaccination induced polyfunctional CD8(+) T cells characterized by coproduction of IFN-γ, TNF-α and IL-2, and this cell phenotype is associated with good viral control. The memory CD8(+) T cells also expressed high levels of CD27 and CD127, which are markers of long-term survival and maintenance of T cell memory. Functionally, the AdIiNS3-vaccinated mice had a significantly increased cytotoxic capacity compared with the AdNS3 group. The AdIiNS3-induced CD8(+) T cells protected mice from infection with recombinant vaccinia virus expressing HCV NS3 of heterologous 1b strains, and studies in knockout mice demonstrated that this protection was mediated primarily through IFN-γ production. On the basis of these promising results, we suggest that this vaccination technology should be evaluated further in the chimpanzee HCV challenge model.


Subject(s)
Adenoviridae/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/immunology , Hepacivirus/immunology , Histocompatibility Antigens Class II/immunology , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/immunology , Adenoviridae/genetics , Animals , Antigens, Differentiation, B-Lymphocyte/administration & dosage , Antigens, Differentiation, B-Lymphocyte/genetics , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Survival/genetics , Cell Survival/immunology , Cytotoxicity Tests, Immunologic/methods , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Vectors/administration & dosage , Hepacivirus/genetics , Hepatitis C/immunology , Hepatitis C/pathology , Hepatitis C/prevention & control , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Immunologic Memory/genetics , Immunophenotyping , Interleukin-7 Receptor alpha Subunit/biosynthesis , Interleukin-7 Receptor alpha Subunit/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Hepatitis Vaccines/administration & dosage , Viral Hepatitis Vaccines/genetics , Viral Nonstructural Proteins/administration & dosage , Viral Nonstructural Proteins/genetics
12.
J Immunol ; 186(2): 856-68, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21149609

ABSTRACT

Chronic graft-versus-host disease (cGVHD) is considered an autoimmune-like disease mediated by donor CD4(+) T cells, but the origin of the autoreactive T cells is still controversial. In this article, we report that the transplantation of DBA/2 donor spleen cells into thymectomized MHC-matched allogeneic BALB/c recipients induced autoimmune-like cGVHD, although not in control syngeneic DBA/2 recipients. The donor-type CD4(+) T cells from the former but not the latter recipients induced autoimmune-like manifestations in secondary allogeneic BALB/c as well as syngeneic DBA/2 recipients. Transfer of donor-type CD4(+) T cells from secondary DBA/2 recipients with disease into syngeneic donor-type or allogeneic host-type tertiary recipients propagated autoimmune-like manifestations in both. Furthermore, TCR spectratyping revealed that the clonal expansion of the autoreactive CD4(+) T cells in cGVHD recipients was initiated by an alloimmune response. Finally, hybridoma CD4(+) T clones derived from DBA/2 recipients with disease proliferated similarly in response to stimulation by syngeneic donor-type or allogeneic host-type dendritic cells. These results demonstrate that the autoimmune-like manifestations in cGVHD can be mediated by a population of donor CD4(+) T cells in transplants that simultaneously recognize Ags presented by both donor and host APCs.


Subject(s)
Autoantigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Graft vs Host Disease/immunology , H-2 Antigens/immunology , Histocompatibility Antigens Class II/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , Autoantibodies/biosynthesis , Autoantigens/administration & dosage , Autoantigens/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/transplantation , Cell Differentiation/genetics , Chronic Disease , Clone Cells , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , Histocompatibility Antigen H-2D , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/genetics , Histocompatibility Testing , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Models, Animal , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/transplantation
13.
J Immunol ; 183(7): 4809-16, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19752238

ABSTRACT

Tissue-specific autoimmune diseases such as type 1 diabetes (T1D) are characterized by T cell-driven pathology. Administration of autoantigenic peptides provides a strategy to selectively target the pathogenic T cell response. Indeed, treatment with beta cell peptides effectively prevents T1D in NOD mice. However, the efficacy of peptide immunotherapy generally wanes as beta cell autoimmunity progresses and islet inflammation increases. With the goal of enhancing the efficacy of peptide immunotherapy, soluble (s)IA(g7)-Ig dimers covalently linked to beta cell autoantigen-derived peptides were tested for the capacity to suppress late preclinical T1D. NOD female mice with established beta cell autoimmunity were vaccinated i.v. with a short course of sIA(g7)-Ig dimers tethered to peptides derived from glutamic acid decarboxylase (GAD)65 (sIA(g7)-pGAD65). Treatment with sIA(g7)-pGAD65 dimers and the equivalent of only approximately 7 microg of native peptide effectively blocked the progression of insulitis and the development of diabetes. Furthermore, suppression of T1D was dependent on beta cell-specific IL-10-secreting CD4+ T cells, although the frequency of GAD65-specific FoxP3-expressing CD4+ T cells was also increased in sIA(g7)-pGAD65 dimer vaccinated NOD mice. These results demonstrate that MHC class II-Ig dimer vaccination is a robust approach to suppress ongoing T cell-mediated autoimmunity, and may provide a superior strategy of adjuvant-free peptide-based immunotherapy to induce immunoregulatory T cells.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Diabetes Mellitus, Type 1/pathology , Dimerization , Epitopes, T-Lymphocyte/genetics , Female , Glutamate Decarboxylase/administration & dosage , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/immunology , Histocompatibility Antigens Class II/genetics , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/transplantation , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
14.
J Immunol ; 181(9): 6584-94, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941249

ABSTRACT

Elevation of CD74 is associated with a number of human cancers, including clear cell renal cell carcinoma (ccRCC). To understand the role of CD74 in the oncogenic process of ccRCC, we ectopically expressed CD74 in human embryonic kidney 293 cells (HEK/CD74) and evaluated its oncogenic potential. Through overexpression of CD74 in HEK293 and Caki-2 cells and down-regulation of CD74 in Caki-1 cells, we show that vascular endothelial growth factor-D (VEGF-D) expression is modified accordingly. A significant, positive correlation between CD74 and VEGF-D is found in human ccRCC tissues (Pearson's correlation, r = 0.65, p < 0.001). In HEK/CD74 xenograft mice, CD74 significantly induced the formation of tumor masses, increased tumor-induced angiogenesis, and promoted cancer cell metastasis. Blockage of VEGF-D expression by small interference RNA resulted in a decrease in cell proliferation, invasion, and cancer cell-induced HUVEC migration enhanced by CD74. Furthermore, we provide evidence that the intracellular signaling cascade responsible for VEGF-D up-regulation by CD74 is both PI3K/AKT- and MEK/ERK-dependent, both of which are associated with NF-kappaB nuclear translocation and DNA-binding activity. These results suggest that VEGF-D is crucial for CD74-induced human renal carcinoma cancer cell tumorigenesis.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/physiology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Gene Expression Regulation, Neoplastic/immunology , Histocompatibility Antigens Class II/physiology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Up-Regulation/immunology , Vascular Endothelial Growth Factor D/biosynthesis , Animals , Antigens, Differentiation, B-Lymphocyte/administration & dosage , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/secondary , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Coculture Techniques , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Histocompatibility Antigens Class II/administration & dosage , Humans , Kidney Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Transplantation, Heterologous , Up-Regulation/genetics , Vascular Endothelial Growth Factor D/genetics , Vascular Endothelial Growth Factor D/physiology
15.
J Immunol ; 181(6): 4371-80, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18768896

ABSTRACT

Mixed chimerism and donor-specific tolerance are achieved in mice receiving 3 Gy of total body irradiation and anti-CD154 mAb followed by allogeneic bone marrow (BM) transplantation. In this model, recipient CD4 cells are critically important for CD8 tolerance. To evaluate the role of CD4 cells recognizing donor MHC class II directly, we used class II-deficient donor marrow and were not able to achieve chimerism unless recipient CD8 cells were depleted, indicating that directly alloreactive CD4 cells were necessary for CD8 tolerance. To identify the MHC class II(+) donor cells promoting this tolerance, we used donor BM lacking certain cell populations or used positively selected cell populations. Neither donor CD11c(+) dendritic cells, B cells, T cells, nor donor-derived IL-10 were critical for chimerism induction. Purified donor B cells induced early chimerism and donor-specific cell-mediated lympholysis tolerance in both strain combinations tested. In contrast, positively selected CD11b(+) monocytes/myeloid cells did not induce early chimerism in either strain combination. Donor cell preparations containing B cells were able to induce early deletion of donor-reactive TCR-transgenic 2C CD8 T cells, whereas those devoid of B cells had reduced activity. Thus, induction of stable mixed chimerism depends on the expression of MHC class II on the donor marrow, but no requisite donor cell lineage was identified. Donor BM-derived B cells induced early chimerism, donor-specific cell-mediated lympholysis tolerance, and deletion of donor-reactive CD8 T cells, whereas CD11b(+) cells did not. Thus, BM-derived B cells are potent tolerogenic APCs for alloreactive CD8 cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Bone Marrow Transplantation/immunology , CD8-Positive T-Lymphocytes/immunology , Clonal Deletion/immunology , Histocompatibility Antigens Class II/immunology , T-Lymphocyte Subsets/immunology , Transplantation Tolerance/immunology , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , B-Lymphocyte Subsets/transplantation , Bone Marrow Transplantation/pathology , CD8-Positive T-Lymphocytes/cytology , Cell Lineage/genetics , Cell Lineage/immunology , Clonal Deletion/genetics , Female , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Radiation Chimera , T-Lymphocyte Subsets/cytology , Transplantation Conditioning , Transplantation Tolerance/genetics
16.
Clin Immunol ; 127(1): 7-13, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18234558

ABSTRACT

We previously showed that immunization of mice with murine fibroblasts transfected with the thyrotropin receptor (TSHR) and a murine major histocompatibility complex (MHC) class II molecule induces immune thyroid disease with the humoral and histological features of human Graves' disease in about 20% of mice. In this model, based on the proliferative response of T cells from hyperthyroid mice to a panel of overlapping TSHR peptides, we now demonstrate that TSHR 121-140 peptide contains an immunodominant T cell epitope. Supporting this conclusion, spleen cells from mice immunized with TSHR 121-140 peptide showed a strong proliferative response to fibroblasts transfected with the TSHR and a murine I-A(k) molecule, but not either alone. Also, intranasal administration of 100 mug of TSHR 121-140 peptide led to suppressed proliferative response of lymph node cells to the peptide. Interestingly, however, administration of this peptide enhanced, rather than suppressed, the frequency and severity of Graves' disease induced by the immunization of the fibroblasts transfected with the TSHR and a murine I-A(k) molecule. Spleen cells from hyperthyroid mice that were pretreated with intranasal peptide tended to produce lesser amounts of IL-4, IL-10 and IFN-gamma than those from normothyroid control mice. Although precise mechanisms of this enhancement remain to be determined, the results suggest that attempts to treat Graves' disease by intranasal administration of an immunodominant TSHR T cell epitope may aggravate, not prevent, the disease.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Graves Disease/immunology , Graves Disease/prevention & control , Immunotherapy/methods , Receptors, Thyrotropin/immunology , Administration, Intranasal , Animals , Cell Proliferation , Cytokines/metabolism , Epitopes, T-Lymphocyte/administration & dosage , Female , Fibroblasts/immunology , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/immunology , Mice , Peptides/administration & dosage , Peptides/immunology , Receptors, Thyrotropin/administration & dosage , T-Lymphocytes/immunology , Transfection
17.
J Immunol ; 180(5): 3339-46, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292559

ABSTRACT

The ideal vaccine induces a potent protective immune response, which should be rapidly induced, long-standing, and of broad specificity. Recombinant adenoviral vectors induce potent Ab and CD8+ T cell responses against transgenic Ags within weeks of administration, and they are among the most potent and versatile Ag delivery vehicles available. However, the impact of chronic infections like HIV and hepatitis C virus underscore the need for further improvements. In this study, we show that the protective immune response to an adenovirus-encoded vaccine Ag can be accelerated, enhanced, broadened, and prolonged by tethering of the rAg to the MHC class II-associated invariant chain (Ii). Thus, adenovirus-vectored vaccines expressing lymphocytic choriomeningitis virus (LCMV)-derived glycoprotein linked to Ii increased the CD4+ and CD8+ T cell stimulatory capacity in vitro and in vivo. Furthermore, mice vaccinated with a single dose of adenovirus-expressing LCMV-derived glycoprotein linked to Ii were protected against lethal virus-induced choriomeningitis, lethal challenge with strains mutated in immunodominant T cell epitopes, and systemic infection with a highly invasive strain. In therapeutic tumor vaccination, the vaccine was as efficient as live LCMV. In comparison, animals vaccinated with a conventional adenovirus vaccine expressing unmodified glycoprotein were protected against systemic infection, but only temporarily against lethal choriomeningitis, and this vaccine was less efficient in tumor therapy.


Subject(s)
Adenoviridae/immunology , Antigens, Differentiation, B-Lymphocyte/administration & dosage , Antigens, Differentiation, B-Lymphocyte/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/immunology , Lymphocytic choriomeningitis virus/immunology , Viral Vaccines/immunology , Adenoviridae/genetics , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cells, Cultured , Coculture Techniques , Genetic Vectors , Histocompatibility Antigens Class II/genetics , Immunity, Cellular/genetics , Lymphocytic choriomeningitis virus/genetics , Lysosomal-Associated Membrane Protein 1/genetics , Lysosomal-Associated Membrane Protein 1/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
18.
J Immunol ; 177(10): 6626-33, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082574

ABSTRACT

The route for presentation of Ag to CD8+ or CD4+ T cells following DNA vaccination is critical for determining outcome, but the pathways involved are unclear. In this study, we compare two different DNA vaccine designs aimed to elicit CD8+ T cell responses against a specific peptide-epitope either by direct- or cross-presentation. Each carries sequences from tetanus toxin (TT) to provide essential CD4+ T cell help. In the first already proven design, the peptide-epitope is fused to the N-terminal domain of fragment C from TT. This appears to act mainly by cross-presentation. In the second design, the peptide-epitope is encoded by a minigene, with induction of Th responses mediated by coexpression of a hybrid invariant chain molecule, incorporating a single determinant from TT (p30) in exchange for class II-associated invariant chain peptide. This design appears to act mainly via direct presentation from transfected APCs. Both vaccines mediated Th-dependent priming of CD8+ T cells in mice, but the kinetics and level of the responses differed markedly, consistent with engagement of distinct pathways of Ag presentation. Importantly, the vaccines could be combined in an alternating prime-boost regime, in either order, generating substantially expanded memory CD8+ T cells, with potent effector function. Taken together, these results demonstrate that vaccination protocols involving different modes of Ag presentation at prime and boost can significantly improve the effectiveness of immunization.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Immunization, Secondary , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Signal Transduction/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Animals , Antigen Presentation/genetics , Antigens, Differentiation, B-Lymphocyte/administration & dosage , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cytotoxicity, Immunologic/genetics , Egg Proteins/administration & dosage , Egg Proteins/genetics , Egg Proteins/immunology , H-2 Antigens/immunology , H-2 Antigens/metabolism , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Immunization, Secondary/methods , Interferon-gamma/biosynthesis , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/genetics , Ovalbumin/immunology , Peptide Fragments/genetics , Signal Transduction/genetics , T-Lymphocytes, Helper-Inducer/immunology , Tetanus Toxin/administration & dosage , Tetanus Toxin/genetics , Tetanus Toxin/immunology , Vaccines, DNA/chemical synthesis , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
19.
J Immunol ; 176(3): 1447-55, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16424172

ABSTRACT

Tumor cells that constitutively express MHC class I molecules and are genetically modified to express MHC class II (MHC II) and costimulatory molecules are immunogenic and have therapeutic efficacy against established primary and metastatic cancers in syngeneic mice and activate tumor-specific human CD4+ T lymphocytes. Previous studies have indicated that these MHC II vaccines enhance immunity by directly activating tumor-specific CD4+ T cells during the immunization process. Because dendritic cells (DCs) are considered to be the most efficient APCs, we have now examined the role of DCs in CD4+ T cell activation by the MHC II vaccines. Surprisingly, we find that DCs are essential for MHC II vaccine immunogenicity; however, they mediate their effect through "cross-dressing." Cross-dressing, or peptide-MHC (pMHC) transfer, involves the generation of pMHC complexes within the vaccine cells, and their subsequent transfer to DCs, which then present the intact, unprocessed complexes to CD4+ T lymphocytes. The net result is that DCs are the functional APCs; however, the immunogenic pMHC complexes are generated by the tumor cells. Because MHC II vaccine cells do not express the MHC II accessory molecules invariant chain and DM, they are likely to load additional tumor Ag epitopes onto MHC II molecules and therefore activate a different repertoire of T cells than DCs. These data further the concept that transfer of cellular material to DCs is important in Ag presentation, and they have direct implications for the design of cancer vaccines.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation/immunology , Lymphoma, B-Cell/immunology , Melanoma, Experimental/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Communication/immunology , Cells, Cultured , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/immunology , Female , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/therapy , Male , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Transgenic , Necrosis , Tumor Cells, Cultured
20.
J Neuroimmunol ; 171(1-2): 8-16, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16271400

ABSTRACT

We have investigated the efficacy of immunization against peptides from predisposing MHC class II molecules in human-compatible adjuvants for ameliorating experimental autoimmune myasthenia gravis (EAMG). C57BL/6 mice were immunized three times with the peptide I-Abetab62-76 in Alum+killed pertussis organisms (PT) prior to two injections with tAChR. The treatment greatly reduced the occurrence and severity of clinical MG relative to controls that received saline/Alum+PT or none. It also reduced antibody and T-cell responses against tAChR. The results have important implications for the possible immunotherapy of MG by targeting disease-associated MHC.


Subject(s)
Histocompatibility Antigens Class II/administration & dosage , Myasthenia Gravis, Autoimmune, Experimental/prevention & control , Pertussis Vaccine/administration & dosage , Vaccination/methods , Action Potentials/physiology , Alum Compounds , Animals , Antibodies/therapeutic use , Antibody Formation , Cell Proliferation/drug effects , Disease Models, Animal , Female , Histocompatibility Antigens Class II/immunology , Humans , Lymph Nodes/cytology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Muscle, Skeletal/physiopathology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/physiopathology , Pertussis Vaccine/immunology , Physical Conditioning, Animal/methods , Radioimmunoassay/methods , Receptors, Cholinergic/immunology , Torpedo
SELECTION OF CITATIONS
SEARCH DETAIL
...