Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Biomaterials ; 220: 119417, 2019 11.
Article in English | MEDLINE | ID: mdl-31419588

ABSTRACT

To improve the efficacy of cancer vaccines we aimed to modulate the suppressive tumor microenvironment. In this study, the potential of intratumoral immune modulation with poly (I:C), Resiquimod (R848) and CCL20 (MIP3α) was explored. Biodegradable polymeric nanoparticles were used as delivery vehicles for slow and sustained release of these drugs in the tumor area and were combined with specific immunotherapy based on therapeutic peptide vaccination in two aggressive murine carcinoma and lymphoma tumor models. Whereas nanoparticle delivery of poly (I:C) or R848 improved therapeutic efficacy, the combination with MIP3α remarkably potentiated the cancer vaccine antitumor effects. The long-term survival increased to 75-100% and the progression free survival nearly doubled on mice with established large carcinoma tumors. The potent adjuvant effects were associated with lymphoid and myeloid population alterations in the tumor and tumor-draining lymph node. In addition to a significant influx of macrophages into the tumor, the phenotype of the suppressor tumor-associated macrophages shifted towards an acute inflammatory phenotype in the tumor-draining lymph node. Overall, these data show that therapeutic cancer vaccines can be potentiated by the combined nanoparticle mediated co-delivery of poly (I:C), R848 and MIP3α, which indicates that a more favorable milieu for cancer fighting immune cells is created for T cells induced by therapeutic cancer vaccines.


Subject(s)
Biocompatible Materials/chemistry , Cancer Vaccines/therapeutic use , Immunologic Factors/administration & dosage , Nanoparticles/chemistry , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Drug Delivery Systems , Endocytosis/drug effects , Imidazoles/administration & dosage , Immunologic Factors/pharmacology , Interleukin-12/biosynthesis , Lymph Nodes/drug effects , Lymph Nodes/pathology , Mice, Inbred C57BL , Poly I-C/administration & dosage , Poly I-C/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Progression-Free Survival , Survival Analysis , Treatment Outcome , Tumor Microenvironment/drug effects , Vaccination
2.
Hum Vaccin Immunother ; 12(4): 1056-69, 2016 04 02.
Article in English | MEDLINE | ID: mdl-26752261

ABSTRACT

Among the emerging subunit vaccines are recombinant protein- and synthetic peptide-based vaccine formulations. However, proteins and peptides have a low intrinsic immunogenicity. A common strategy to overcome this is to co-deliver (an) antigen(s) with (an) immune modulator(s) by co-encapsulating them in a particulate delivery system, such as poly(lactic-co-glycolic acid) (PLGA) particles. Particulate PLGA formulations offer many advantages for antigen delivery as they are biocompatible and biodegradable; can protect the antigens from degradation and clearance; allow for co-encapsulation of antigens and immune modulators; can be targeted to antigen presenting cells; and their particulate nature can increase uptake and cross-presentation by mimicking the size and shape of an invading pathogen. In this review we discuss the pros and cons of using PLGA particulate formulations for subunit vaccine delivery and provide an overview of formulation parameters that influence their adjuvanticity and the ensuing immune response.


Subject(s)
Lactic Acid , Polyglycolic Acid , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Antigens/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cross-Priming , Dendritic Cells/immunology , Drug Delivery Systems , Humans , Immunity, Cellular , Immunogenicity, Vaccine , Nanoparticles , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Vaccines, Subunit/adverse effects , Vaccines, Subunit/chemistry
3.
PLoS One ; 10(6): e0127969, 2015.
Article in English | MEDLINE | ID: mdl-26046664

ABSTRACT

Currently licensed influenza vaccines mainly induce antibodies against highly variable epitopes. Due to antigenic drift, protection is subtype or strain-specific and regular vaccine updates are required. In case of antigenic shifts, which have caused several pandemics in the past, completely new vaccines need to be developed. We set out to develop a vaccine that provides protection against a broad range of influenza viruses. Therefore, highly conserved parts of the influenza A virus (IAV) were selected of which we constructed antibody and T cell inducing peptide-based vaccines. The B epitope vaccine consists of the highly conserved HA2 fusion peptide and M2e peptide coupled to a CD4 helper epitope. The T epitope vaccine comprises 25 overlapping synthetic long peptides of 26-34 amino acids, thereby avoiding restriction for a certain MHC haplotype. These peptides are derived from nucleoprotein (NP), polymerase basic protein 1 (PB1) and matrix protein 1 (M1). C57BL/6 mice, BALB/c mice, and ferrets were vaccinated with the B epitopes, 25 SLP or a combination of both. Vaccine-specific antibodies were detected in sera of mice and ferrets and vaccine-specific cellular responses were measured in mice. Following challenge, both mice and ferrets showed a reduction of virus titers in the lungs in response to vaccination. Summarizing, a peptide-based vaccine directed against conserved parts of influenza virus containing B and T cell epitopes shows promising results for further development. Such a vaccine may reduce disease burden and virus transmission during pandemic outbreaks.


Subject(s)
Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Influenza Vaccines/immunology , Lung/virology , Vaccines, Subunit/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Databases, Factual , Dogs , Female , Ferrets , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H5N1 Subtype/metabolism , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Vaccines, Subunit/chemical synthesis , Vaccines, Subunit/chemistry , Viral Load , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/immunology
4.
Vaccine ; 33(7): 847-54, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25576216

ABSTRACT

Poly(lactic-co-glycolic acid) (PLGA) particles have been extensively studied as biodegradable delivery system to improve the potency and safety of protein-based vaccines. In this study we analyzed how the size of PLGA particles, and hence their ability to be engulfed by dendritic cells (DC), affects the type and magnitude of the immune response in comparison to sustained release from a local depot. PLGA microparticles (MP, volume mean diameter≈112 µm) and nanoparticles (NP, Z-average diameter≈350 nm) co-encapsulating ovalbumin (OVA) and poly(I:C), with comparable antigen (Ag) release characteristics, were prepared and characterized. The immunogenicity of these two distinct particulate vaccines was evaluated in vitro and in vivo. NP were efficiently taken up by DC and greatly facilitated MHC I Ag presentation in vitro, whereas DC cultured in the presence of MP failed to internalize significant amounts of Ag and hardly showed MHC I Ag presentation. Vaccination of mice with NP resulted in significantly better priming of Ag-specific CD8(+) T cells compared to MP and OVA emulsified with incomplete Freund's adjuvant (IFA). Moreover, NP induced a balanced TH1/TH2-type antibody response, compared to vaccinations with IFA which stimulated a predominant TH2-type response, whereas MP failed to increase antibody titers. In conclusion, we postulate that particle internalization is of crucial importance and therefore particulate vaccines should be formulated in the nano- but not micro-size range to achieve efficient uptake, significant MHC class I cross-presentation and effective T and B cell responses.


Subject(s)
Dendritic Cells/immunology , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Vaccines/immunology , Animals , Antibody Formation/immunology , CD8-Positive T-Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Polylactic Acid-Polyglycolic Acid Copolymer
5.
Eur J Pharm Biopharm ; 83(3): 338-45, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23201055

ABSTRACT

Overlapping synthetic long peptides (SLPs) hold great promise for immunotherapy of cancer. Poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) are being developed as delivery systems to improve the potency of peptide-based therapeutic cancer vaccines. Our aim was to optimize PLGA NP for SLP delivery with respect to encapsulation and release, using OVA24, a 24-residue long synthetic antigenic peptide covering a CTL epitope of ovalbumin (SIINFEKL), as a model antigen. Peptide-loaded PLGA NPs were prepared by a double emulsion/solvent evaporation technique. Using standard conditions (acidic inner aqueous phase), we observed that either encapsulation was very low (1-30%), or burst release extremely high (>70%) upon resuspension of NP in physiological buffers. By adjusting formulation and process parameters, we uncovered that the pH of the first emulsion was critical to efficient encapsulation and controlled release. In particular, an alkaline inner aqueous phase resulted in circa 330 nm sized NP with approximately 40% encapsulation efficiency and low (<10%) burst release. These NP showed enhanced MHC class I restricted T cell activation in vitro when compared to high-burst releasing NP and soluble OVA24, proving that efficient entrapment of the antigen is crucial to induce a potent cellular immune response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lactic Acid/chemistry , Lymphocyte Activation , Nanoparticles , Peptides/chemistry , Polyglycolic Acid/chemistry , Amino Acid Sequence , Molecular Sequence Data , Polylactic Acid-Polyglycolic Acid Copolymer
6.
Ann N Y Acad Sci ; 1103: 192-5, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17376840

ABSTRACT

Type 1 diabetes (T1D) is a multifactorial disease characterized by the infiltration and subsequent destruction of the pancreatic insulin-producing beta cells by autoreactive T cells. CD8(+) T cells play an essential role in this beta cell destruction. However, little is known about the target antigens of CD8(+) T cells in human T1D patients. The aim of this study was to assess whether an epitope derived from the islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP), IGRP(265-273,) which has recently been identified as a target in non-obese diabetic (NOD) mice and is fully homologous to the human epitope, is a target of human diabetogenic CD8(+) T cells. We isolated a human CD8 T cell clone against this epitope, which confirms that this IGRP epitope is shared across species.


Subject(s)
Autoantigens/immunology , CD8 Antigens/immunology , Diabetes Mellitus, Type 1/immunology , Glucose-6-Phosphatase/immunology , Islets of Langerhans/immunology , Proteins/immunology , Animals , Disease Models, Animal , Epitopes/immunology , Humans , Mice
7.
Leukemia ; 20(10): 1738-50, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16932347

ABSTRACT

For immunotherapy of residual disease in patients with Philadelphia-positive leukemias, the BCR-ABL fusion regions are attractive disease-specific T-cell targets. We analyzed these regions for the prevalence of cytotoxic T lymphocyte (CTL) epitopes by an advanced reverse immunology procedure. Seventeen novel BCR-ABL fusion peptides were identified to bind efficiently to the human lymphocyte antigen (HLA)-A68, HLA-B51, HLA-B61 or HLA-Cw4 HLA class I molecules. Comprehensive enzymatic digestion analysis showed that 10 out of the 28 HLA class I binding fusion peptides were efficiently excised after their C-terminus by the proteasome, which is an essential requirement for efficient cell surface expression. Therefore, these peptides are prime vaccine candidates. The other peptides either completely lacked C-terminal liberation or were only inefficiently excised by the proteasome, rendering them inappropriate or less suitable for inclusion in a vaccine. CTL raised against the properly processed HLA-B61 epitope AEALQRPVA from the BCR-ABL e1a2 fusion region, expressed in acute lymphoblastic leukemia (ALL), specifically recognized ALL tumor cells, proving cell surface presentation of this epitope, its applicability for immunotherapy and underlining the accuracy of our epitope identification strategy. Our study provides a reliable basis for the selection of optimal peptides to be included in immunotherapeutic BCR-ABL vaccines against leukemia.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Fusion Proteins, bcr-abl/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Amino Acid Sequence , Cell Line, Tumor , Epitope Mapping/methods , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , HLA-A Antigens/immunology , HLA-A Antigens/metabolism , HLA-A2 Antigen , HLA-B Antigens/immunology , HLA-B Antigens/metabolism , HLA-B51 Antigen , HLA-C Antigens/immunology , HLA-C Antigens/metabolism , Humans , Immunotherapy/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding/immunology
8.
Eur J Immunol ; 31(12): 3602-11, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11745380

ABSTRACT

Bacterial antigens recognized by CD8(+) T cells in the context of MHC class I are thought to play a crucial role in protection against pathogenic intracellular bacteria. Here, we demonstrate the induction of HLA-A*0201-restricted CD8(+) T cell responses against six new high-affinity HLA-A*0201-binding CTL epitopes, encoded within an immunodominant and highly conserved antigen of Mycobacteria, the heat shock protein 65 (hsp65). One of these epitopes, Mhsp65(9(369)), is identical in a large number of pathogenic bacteria, and is recognized in a CD8-independent fashion. Mhsp65(9(369)) could be presented by either mycobacterial hsp65-pulsed target cells or BCG-infected macrophages. Interestingly, T cells specific for this epitope did not recognize the corresponding human hsp65 homologue, probably due to structural differences as revealed by modeling studies. Furthermore, in vitro proteasome digestion analyses show that, whereas the mycobacterial hsp65 epitope is efficiently generated, the human hsp65 homologue is not, thus avoiding the induction of autoreactivity. Collectively, these findings describe high-affinity HLA class I-binding epitopes that are naturally processed and are recognized efficiently by MHC class I-restricted CD8(+) T cells, providing a rational basis for the development of subunit vaccine strategies against tuberculosis and other intracellular infectious diseases.


Subject(s)
Bacterial Proteins , Chaperonins/immunology , Epitopes, T-Lymphocyte , HLA-A Antigens/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Antigen Presentation , Cell Line , Chaperonin 60 , Chaperonins/chemistry , Cysteine Endopeptidases/physiology , Female , HLA-A2 Antigen , Humans , Immunization , Male , Mice , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Multienzyme Complexes/physiology , Mycobacterium/immunology , Proteasome Endopeptidase Complex , Vaccines, DNA/immunology
9.
J Cell Biol ; 155(1): 53-63, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11581285

ABSTRACT

Immature dendritic cells (DCs) sample their environment for antigens and after stimulation present peptide associated with major histocompatibility complex class II (MHC II) to naive T cells. We have studied the intracellular trafficking of MHC II in cultured DCs. In immature cells, the majority of MHC II was stored intracellularly at the internal vesicles of multivesicular bodies (MVBs). In contrast, DM, an accessory molecule required for peptide loading, was located predominantly at the limiting membrane of MVBs. After stimulation, the internal vesicles carrying MHC II were transferred to the limiting membrane of the MVB, bringing MHC II and DM to the same membrane domain. Concomitantly, the MVBs transformed into long tubular organelles that extended into the periphery of the cells. Vesicles that were formed at the tips of these tubules nonselectively incorporated MHC II and DM and presumably mediated transport to the plasma membrane. We propose that in maturing DCs, the reorganization of MVBs is fundamental for the timing of MHC II antigen loading and transport to the plasma membrane.


Subject(s)
Antigen Presentation , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Transport Vesicles/metabolism , Animals , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Endocytosis/physiology , Histocompatibility Antigens Class II/metabolism , Lipopolysaccharides/pharmacology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Protein Transport , Up-Regulation
10.
J Exp Med ; 194(5): 657-67, 2001 Sep 03.
Article in English | MEDLINE | ID: mdl-11535633

ABSTRACT

Dendritic cells (DCs) play a central role in the immune system as they drive activation of T lymphocytes by cognate interactions. However, as DCs express high levels of major histocompatibility complex class I, this intimate contact may also result in elimination of DCs by activated cytotoxic T lymphocytes (CTLs) and thereby limit induction of immunity. We show here that immature DCs are indeed susceptible to CTL-induced killing, but become resistant upon maturation with anti-CD40 or lipopolysaccharide. Protection is achieved by expression of serine protease inhibitor (SPI)-6, a member of the serpin family that specifically inactivates granzyme B and thereby blocks CTL-induced apoptosis. Anti-CD40 and LPS-induced SPI-6 expression is sustained for long periods of time, suggesting a role for SPI-6 in the longevity of DCs. Importantly, T helper 1 cells, which mature DCs and boost CTL immunity, induce SPI-6 expression and subsequent DC resistance. In contrast, T helper 2 cells neither induce SPI-6 nor convey protection, despite the fact that they trigger DC maturation with comparable efficiency. Our data identify SPI-6 as a novel marker for DC function, which protects DCs against CTL-induced apoptosis.


Subject(s)
Apoptosis/immunology , Dendritic Cells/immunology , Receptors, Antigen, T-Cell/immunology , Serine Proteinase Inhibitors/metabolism , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Apoptosis/drug effects , CD40 Antigens/immunology , CD40 Antigens/physiology , Cells, Cultured , Dendritic Cells/cytology , Flow Cytometry , Granzymes , Humans , Kinetics , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Ovalbumin/immunology , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/genetics , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/genetics
11.
J Exp Med ; 193(1): 73-88, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11136822

ABSTRACT

We report the efficient identification of four human histocompatibility leukocyte antigen (HLA)-A(*)0201-presented cytotoxic T lymphocyte (CTL) epitopes in the tumor-associated antigen PRAME using an improved "reverse immunology" strategy. Next to motif-based HLA-A(*)0201 binding prediction and actual binding and stability assays, analysis of in vitro proteasome-mediated digestions of polypeptides encompassing candidate epitopes was incorporated in the epitope prediction procedure. Proteasome cleavage pattern analysis, in particular determination of correct COOH-terminal cleavage of the putative epitope, allows a far more accurate and selective prediction of CTL epitopes. Only 4 of 19 high affinity HLA-A(*)0201 binding peptides (21%) were found to be efficiently generated by the proteasome in vitro. This approach avoids laborious CTL response inductions against high affinity binding peptides that are not processed and limits the number of peptides to be assayed for binding. CTL clones induced against the four identified epitopes (VLDGLDVLL, PRA(100-108); SLYSFPEPEA, PRA(142-151); ALYVDSLFFL, PRA(300-309); and SLLQHLIGL, PRA(425-433)) lysed melanoma, renal cell carcinoma, lung carcinoma, and mammary carcinoma cell lines expressing PRAME and HLA-A(*)0201. This indicates that these epitopes are expressed on cancer cells of diverse histologic origin, making them attractive targets for immunotherapy of cancer.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/metabolism , Cysteine Endopeptidases/metabolism , HLA-A Antigens/metabolism , Multienzyme Complexes/metabolism , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Antigens, Neoplasm/genetics , Base Sequence , Cell Line, Transformed , Cytotoxicity, Immunologic , DNA Primers/genetics , Epitopes/genetics , Epitopes/metabolism , Humans , Molecular Sequence Data , Proteasome Endopeptidase Complex , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Tumor Cells, Cultured
12.
Cancer Res ; 60(22): 6427-33, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11103809

ABSTRACT

Efficient loading of MHC class II molecules with a T helper epitope of choice can be achieved through genetic exchange of the MHC class II-associated invariant chain peptide (CLIP) sequence with a sequence encoding the helper peptide. We have now used this method to engineer a cellular vaccine that continuously expresses a tumor-specific helper epitope in a defined costimulatory context. We provide evidence (a) that this cellular vaccine induces peptide-specific helper T cells in vivo that are functional in protecting mice from challenge with a highly aggressive tumor, (b) that this vaccine can directly prime tumor-specific helper T cells in vivo, and (c) that this cellular vaccine is superior compared with similar cells loaded with synthetic T helper peptide in inducing tumor protection. In conclusion, cellular vaccines for activation of antigen-specific helper T cells can be greatly improved by the introduction of invariant chain constructs containing a T helper epitope by class II-associated invariant chain peptide exchange.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/physiology , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Neoplasm/genetics , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Female , Histocompatibility Antigens Class II/genetics , Lymphocyte Activation/immunology , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Retroviridae Proteins, Oncogenic/immunology , Transfection , Vaccination , Viral Envelope Proteins/immunology
14.
Curr Opin Immunol ; 12(5): 576-82, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11007362

ABSTRACT

After studies in preclinical mouse models, the efficacy and safety of tumor-specific vaccination strategies is currently being evaluated in cancer patients. The first wave of clinical trials has shown that in general such vaccination strategies are safe. However examples of clinical responses, especially in conjunction with vaccine-induced immune responses, are still scarce. The fact that most trials have so far been performed with end-stage cancer patients can largely account for this deficit. Greater efficacy of anticancer vaccines is expected in patients with less-progressed disease. In addition, the detection of both natural and vaccine-induced T cell immunity needs further improvement.


Subject(s)
Antigens/immunology , Cancer Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes , HLA Antigens/immunology , Humans , T-Lymphocytes, Cytotoxic/immunology , Vaccination
15.
J Exp Med ; 192(4): 483-94, 2000 Aug 21.
Article in English | MEDLINE | ID: mdl-10952718

ABSTRACT

The proteasome is the principal provider of major histocompatibility complex (MHC) class I-presented peptides. Interferon (IFN)-gamma induces expression of three catalytically active proteasome subunits (LMP2, LMP7, and MECL-1) and the proteasome-associated activator PA28. These molecules are thought to optimize the generation of MHC class I-presented peptides. However, known information on their contribution in vivo is very limited. Here, we examined the antigen processing of two murine leukemia virus-encoded cytotoxic T lymphocyte (CTL) epitopes in murine cell lines equipped with a tetracycline-controlled, IFN-gamma-independent expression system. We thus were able to segregate the role of the immunosubunits from the role of PA28. The presence of either immunosubunits or PA28 did not alter the presentation of a subdominant murine leukemia virus (MuLV)-derived CTL epitope. However, the presentation of the immunodominant MuLV-derived epitope was markedly enhanced upon induction of each of these two sets of genes. Thus, the IFN-gamma-inducible proteasome subunits and PA28 can independently enhance antigen presentation of some CTL epitopes. Our data show that tetracycline-regulated expression of PA28 increases CTL epitope generation without affecting the 20S proteasome composition or half-life. The differential effect of these IFN-gamma-inducible proteins on MHC class I processing may have a decisive influence on the quality of the CTL immune response.


Subject(s)
Acetylcysteine/analogs & derivatives , Antigen Presentation , Cysteine Endopeptidases/metabolism , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Multienzyme Complexes/metabolism , Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , ATP-Binding Cassette Transporters/metabolism , Acetylcysteine/pharmacology , Animals , Autoantigens , Blotting, Western , Cell Cycle Proteins , Cell Line , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/immunology , Cysteine Proteinase Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Interferon-gamma/metabolism , Leukemia Virus, Murine/immunology , Mice , Mice, Inbred C57BL , Multienzyme Complexes/immunology , Precipitin Tests , Proteasome Endopeptidase Complex , Proteins/genetics , Proteins/immunology , Rats , Sulfones/pharmacology , Tetracycline/pharmacology
17.
J Exp Med ; 192(1): 145-50, 2000 Jul 03.
Article in English | MEDLINE | ID: mdl-10880536

ABSTRACT

The well defined, immature murine dendritic cell (DC) line D1 was used to study the role of DC maturation in CTL induction in vitro and in vivo. Maturation of D1 cells, characterized by markedly increased expression of MHC and costimulatory molecules, was induced by incubation with lipopolysaccharide, agonistic CD40 antibody, or specific CD4(+) T helper (Th) cells. Activated, but not immature, D1 cells efficiently primed alloreactive T cell responses in vitro. Similarly, priming of CTL immunity in vivo in CD4-depleted mice was only observed if these mice were immunized with activated D1 cells. This study provides formal evidence that activation of DCs, induced by Th-independent as well as Th-dependent stimuli, is essential for efficient induction of CTL responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies/pharmacology , CD40 Antigens/immunology , Cell Line , Coculture Techniques , Dendritic Cells/drug effects , Female , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Major Histocompatibility Complex , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Th1 Cells/immunology
18.
J Immunol ; 165(2): 869-77, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10878361

ABSTRACT

C57BL/6 mice generate a vigorous H-2Db-restricted CTL response against murine leukemia virus (MuLV)-induced tumors. For many years it has been suggested that this response is directed to an MuLV-encoded peptide as well as to a nonviral tumor-associated peptide. Recently, a peptide from the leader sequence of gag was demonstrated to be the MuLV-derived epitope. Here we describe the molecular identification of the tumor-associated epitope. Furthermore, we show that the CTL response against this epitope can restrict the outgrowth of MuLV-induced tumors in vivo. The epitope is selectively presented by the MuLV-induced T cell tumors RBL-5, RMA, and MBL-2 as well as by the chemically induced T cell lymphoma EL-4. Intriguingly, these tumors share expression of the newly identified epitope because they represent variants of the same clonal tumor cell line, as evident from sequencing of the TCR alpha- and beta-chains, which proved to be identical. Our research shows that all sources of RBL-5, RMA, RMA-S, MBL-2, and EL-4 tumors are derived from a single tumor line, most likely EL-4.


Subject(s)
Antigen Presentation , Epitopes, T-Lymphocyte/metabolism , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Adoptive Transfer , Animals , Binding Sites/immunology , Cancer Vaccines/administration & dosage , Cell Lineage/genetics , Cell Lineage/immunology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/isolation & purification , Friend murine leukemia virus , Gene Expression Regulation/immunology , Genes, T-Cell Receptor beta , Leukemia, Experimental/immunology , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Lymphoma, T-Cell/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Moloney murine leukemia virus , Oligopeptides/administration & dosage , Oligopeptides/chemical synthesis , Oligopeptides/immunology , Oligopeptides/metabolism , Rauscher Virus , Sequence Analysis, Protein , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/transplantation , Thymoma/immunology , Thymoma/metabolism , Thymoma/pathology , Tumor Cells, Cultured/transplantation , Tumor Virus Infections/immunology , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology
19.
J Immunol ; 164(9): 4500-6, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10779750

ABSTRACT

Proteasomes are the major source for the generation of peptides bound by MHC class I molecules. To study the functional relevance of the IFN-gamma-inducible proteasome subunits low molecular mass protein 2 (LMP2), LMP7, and mouse embryonal cell (MEC) ligand 1 in Ag processing and concomitantly that of immunoproteasomes, we established the tetracycline-regulated mouse cell line MEC217, allowing the titrable formation of immunoproteasomes. Infection of MEC217 cells with Adenovirus type 5 (Ad5) and analysis of Ag presentation with Ad5-specific CTL showed that cells containing immunoproteasomes processed the viral early 1B protein (E1B)-derived epitope E1B192-200 with increased efficiency, thus allowing a faster detection of viral entry in induced cells. Importantly, optimal CTL activation was already achieved at submaximal immunosubunit expression. In contrast, digestion of E1B-polypeptide with purified proteasomes in vitro yielded E1B192-200 at quantities that were proportional to the relative contents of immunosubunits. Our data provide evidence that the IFN-gamma-inducible proteasome subunits, when present at relatively low levels as at initial stages of infection, already increase the efficiency of antigenic peptide generation and thereby enhance MHC class I Ag processing in infected cells.


Subject(s)
Adenoviruses, Human/immunology , Antigen Presentation , Cysteine Endopeptidases/immunology , Cysteine Endopeptidases/metabolism , Epitopes, T-Lymphocyte/metabolism , Histocompatibility Antigens Class I/metabolism , Multienzyme Complexes/immunology , Multienzyme Complexes/metabolism , T-Lymphocytes, Cytotoxic/enzymology , T-Lymphocytes, Cytotoxic/immunology , Adenoviruses, Human/genetics , Adjuvants, Immunologic/physiology , Amino Acid Sequence , Animals , Antigen Presentation/drug effects , Antigen Presentation/genetics , Cell Line , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/physiology , Dose-Response Relationship, Immunologic , Enzyme Induction/drug effects , Enzyme Induction/genetics , Enzyme Induction/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Multienzyme Complexes/biosynthesis , Multienzyme Complexes/physiology , Peptide Biosynthesis/immunology , Proteasome Endopeptidase Complex , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology , Tetracycline/pharmacology , Transfection , Tumor Cells, Cultured
20.
J Immunol ; 164(4): 1898-905, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657639

ABSTRACT

CTL directed against the Moloney murine leukemia virus (MuLV) epitope SSWDFITV recognize Moloney MuLV-induced tumor cells, but do not recognize cells transformed by the closely related Friend MuLV. The potential Friend MuLV epitope has strong sequence homology with Moloney MuLV and only differs in one amino acid within the CTL epitope and one amino acid just outside the epitope. We now show that failure to recognize Friend MuLV-transformed tumor cells is based on a defect in proteasome-mediated processing of the Friend epitope which is due to a single amino acid substitution (N-->D) immediately flanking the C-terminal anchor residue of the epitope. Proteasome-mediated digestion analysis of a synthetic 26-mer peptide derived from the Friend sequence shows that cleavage takes place predominantly C-terminal of D, instead of V as is the case for the Moloney MuLV sequence. Therefore, the C terminus of the epitope is not properly generated. Epitope-containing peptide fragments extended with an additional C-terminal D are not efficiently translocated by TAP and do not show significant binding affinity to MHC class I-Kb molecules. Thus, a potential CTL epitope present in the Friend virus sequence is not properly processed and presented because of a natural flanking aspartic acid that obliterates the correct C-terminal cleavage site. This constitutes a novel way to subvert proteasome-mediated generation of proper antigenic peptide fragments.


Subject(s)
Amino Acid Substitution/immunology , Cysteine Endopeptidases/metabolism , Epitopes, T-Lymphocyte/metabolism , Multienzyme Complexes/metabolism , Peptide Fragments/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , ATP-Binding Cassette Transporters/metabolism , Amino Acid Sequence , Animals , Antigen Presentation , Friend murine leukemia virus/immunology , H-2 Antigens/metabolism , HeLa Cells , Humans , Hydrolysis , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Moloney murine leukemia virus/immunology , Peptide Fragments/immunology , Proteasome Endopeptidase Complex , Protein Precursors/immunology , Protein Precursors/metabolism , Rauscher Virus/immunology , T-Lymphocytes, Cytotoxic/enzymology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...