Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Cell Mol Med ; 28(13): e18470, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38963257

ABSTRACT

Recombinant antibodies (Abs) are an integral modality for the treatment of multiple tumour malignancies. Since the Food and Drug Administration (FDA) approval of rituximab as the first monoclonal antibody (mAb) for cancer treatment, several mAbs and antibody (Ab)-based therapies have been approved for the treatment of solid tumour malignancies and other cancers. These Abs function by either blocking oncogenic pathways or angiogenesis, modulating immune response, or by delivering a conjugated drug. The use of Ab-based therapy in cancer patients who could benefit from the treatment, however, is still limited by associated toxicity profiles which may stem from biological features and processes related to target binding, alongside biochemical and/or biophysical characteristics of the therapeutic Ab. A significant immune-related adverse event (irAE) associated with Ab-based therapies is cytokine release syndrome (CRS), characterized by the development of fever, rash and even marked, life-threatening hypotension, and acute inflammation with secondary to systemic uncontrolled increase in a range of pro-inflammatory cytokines. Here, we review irAEs associated with specific classes of approved, Ab-based novel cancer immunotherapeutics, namely immune checkpoint (IC)-targeting Abs, bispecific Abs (BsAbs) and Ab-drug-conjugates (ADCs), highlighting the significance of harmonization in preclinical assay development for safety assessment of Ab-based biotherapeutics as an approach to support and refine clinical translation.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/therapeutic use , Immunotherapy/adverse effects , Immunotherapy/methods , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use
2.
FASEB J ; 37(6): e22995, 2023 06.
Article in English | MEDLINE | ID: mdl-37219526

ABSTRACT

Immuno-oncology (IO)-based therapies such as checkpoint inhibitors, bi-specific antibodies, and CAR-T-cell therapies have shown significant success in the treatment of several cancer indications. However, these therapies can result in the development of severe adverse events, including cytokine release syndrome (CRS). Currently, there is a paucity of in vivo models that can evaluate dose-response relationships for both tumor control and CRS-related safety issues. We tested an in vivo PBMC humanized mouse model to assess both treatment efficacy against specific tumors and the concurrent cytokine release profiles for individual human donors after treatment with a CD19xCD3 bispecific T-cell engager (BiTE). Using this model, we evaluated tumor burden, T-cell activation, and cytokine release in response to bispecific T-cell-engaging antibody in humanized mice generated with different PBMC donors. The results show that PBMC engrafted NOD-scid Il2rgnull mice lacking expression of mouse MHC class I and II (NSG-MHC-DKO mice) and implanted with a tumor xenograft predict both efficacy for tumor control by CD19xCD3 BiTE and stimulated cytokine release. Moreover, our findings indicate that this PBMC-engrafted model captures variability among donors for tumor control and cytokine release following treatment. Tumor control and cytokine release were reproducible for the same PBMC donor in separate experiments. The PBMC humanized mouse model described here is a sensitive and reproducible platform that identifies specific patient/cancer/therapy combinations for treatment efficacy and development of complications.


Subject(s)
Leukocytes, Mononuclear , T-Lymphocytes , Humans , Animals , Mice , Mice, Inbred NOD , Treatment Outcome , Cytokine Release Syndrome , Cytokines , Disease Models, Animal , Mice, Knockout , Mice, SCID
3.
J Vis Exp ; (199)2023 09 15.
Article in English | MEDLINE | ID: mdl-37782084

ABSTRACT

New immunostimulatory antibody drugs designed to either directly stimulate specific immune cells or indirectly enhance the immune response by blocking or activating an endogenous regulator of the immune system have the potential to cause serious immune-related adverse events such as cytokine release syndrome (CRS). It is, therefore crucial to assess the safety profile of such drugs with a combination of in vivo and in vitro experiments before first-in-human dose administration. Cytokine release assays (CRAs), where the proposed antibody therapeutic is co-cultured with human immune cells (such as peripheral blood mononuclear cells (PBMCs), whole blood, or otherwise) and the amount of inflammatory cytokine produced is measured, are critical for hazard identification. However, different labs using different control antibodies can threaten the harmonization of CRAs, and clinically relevant controls (such as TGN1412) can be difficult to source, which can lead to less accurate or reliable results or data which are difficult to compare between laboratories. The inclusion of positive and negative controls in a CRA can ensure the accuracy and reliability of the results. The National Institute for Biological Standards and Control (NIBSC) has produced a panel of lyophilized antibody controls intended for use in various CRA platforms to harmonize results across various laboratories and assay methods. A set of three different positive control antibodies include anti-CD52, anti-CD3, and anti-CD28 superagonist (SA), which are known to induce dose-dependent CRS in patients. Each antibody is provided with an isotype-matched negative control antibody. This panel of reference reagents has previously been shown to have good inter-lab reproducibility and are suitable controls to increase the confidence and robustness of safety data from a variety of CRA platforms.


Subject(s)
Antibodies, Monoclonal , Leukocytes, Mononuclear , Humans , Antibodies, Monoclonal/pharmacology , Reproducibility of Results , Cytokines , CD28 Antigens
4.
Blood ; 115(10): 1949-57, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20065291

ABSTRACT

There is a high demand for the development of adjuvants that induce cytotoxic T lymphocytes, which are crucial for the elimination of intracellular pathogens and tumor cells. Toll-like receptor (TLR) agonists are prime candidates to fulfill this role because they induce innate immune activation and promote adaptive immune responses. The successful application of the TLR7 agonist R837 for treatment of basal cell carcinoma shows the potential for exploiting this pathway in tumor immunotherapy. Imidazoquinolines like R837 and stimulatory ssRNA oligonucleotides both trigger TLR7-mediated immune activation, but little is known about their comparative ability to promote immunity induction. We investigated differences in innate immune activation and adjuvant activity between the imidazoquinoline R848 and the ssRNA TLR7 agonist polyUs21. In contrast to R848, polyUs21 induced detectable levels of intracellular interferon-alpha (IFN-alpha) in plasmacytoid dendritic cells (PDCs). In immunization studies, only polyUs21 led to robust priming of type 1 T helper cells and cytotoxic T lymphocytes, and it was more efficient in inducing antitumor immunity than R848. Notably, exogenous IFN-alpha augmented the adjuvant activity of R848, whereas depletion of PDC abrogated the adjuvanticity of polyUs21. This study, therefore, identifies sufficient IFN-alpha production by PDC as an important determinant of vaccine efficacy.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/metabolism , Interferon Type I/physiology , Membrane Glycoproteins/agonists , Toll-Like Receptor 7/agonists , Aminoquinolines/pharmacology , Aminoquinolines/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Imidazoles/therapeutic use , Imiquimod , Interferon Type I/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Quinolines/pharmacology , RNA/pharmacology , Receptor, Interferon alpha-beta/genetics , Tumor Cells, Cultured
5.
Sci Rep ; 12(1): 18694, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333445

ABSTRACT

SARS-CoV-2 exhibits a diverse host species range with variable outcomes, enabling differential host susceptibility studies to assess suitability for pre-clinical countermeasure and pathogenesis studies. Baseline virological, molecular and pathological outcomes were determined among multiple species-one Old World non-human primate (NHP) species (cynomolgus macaques), two New World NHP species (red-bellied tamarins; common marmosets) and Syrian hamsters-following single-dose, atraumatic intranasal administration of SARS-CoV-2/Victoria-01. After serial sacrifice 2, 10 and 28-days post-infection (dpi), hamsters and cynomolgus macaques displayed differential virus biodistribution across respiratory, gastrointestinal and cardiovascular systems. Uniquely, New World tamarins, unlike marmosets, exhibited high levels of acute upper airway infection, infectious virus recovery associated with mild lung pathology representing a host previously unrecognized as susceptible to SARS-CoV-2. Across all species, lung pathology was identified post-clearance of virus shedding (antigen/RNA), with an association of virus particles within replication organelles in lung sections analysed by electron microscopy. Disrupted cell ultrastructure and lung architecture, including abnormal morphology of mitochondria 10-28 dpi, represented on-going pathophysiological consequences of SARS-CoV-2 in predominantly asymptomatic hosts. Infection kinetics and host pathology comparators using standardized methodologies enables model selection to bridge differential outcomes within upper and lower respiratory tracts and elucidate longer-term consequences of asymptomatic SARS-CoV-2 infection.


Subject(s)
COVID-19 , SARS-CoV-2 , Cricetinae , Animals , Tissue Distribution , Administration, Intranasal , Disease Models, Animal , Lung/pathology , Mesocricetus , Macaca fascicularis
6.
Article in English | MEDLINE | ID: mdl-34095644

ABSTRACT

Accurate determination of cellular subsets that secrete particular cytokine(s) is a significant parameter for functional characterization of an immunological response. The present study was conducted to develop a method for simultaneous measurement of intracellular cytokine positive CD4 and CD8 positive T lymphocytes in a single tube, with a no-wash protocol. We report here the development of a simplified, rapid procedure for precise enumeration of cytokine positive T lymphocytes using BD Trucount tubes. This single step protocol for accurate enumeration of cytokine positive T lymphocytes, will allow for better characterization of immune cell phenotype and function.

7.
Pharmacology ; 83(5): 275-86, 2009.
Article in English | MEDLINE | ID: mdl-19321962

ABSTRACT

BACKGROUND: Intracellular cyclic adenosine monophosphate (cAMP) in inflammatory cells and airway smooth muscle is critical to the modulation of inflammatory response generation. Phosphodiesterase 4 (PDE4), an enzyme that catalyzes cAMP degradation, is therefore being actively explored as a molecular target for the treatment of airway inflammation, particularly asthma and chronic obstructive pulmonary disease. The field has undergone major advances in optimizing generation of compounds with a safe therapeutic margin; however, most PDE4 inhibitors tested so far have unacceptable side effects, particularly nausea and vomiting. METHODS: We evaluated NIS-62949 in a wide range of in vitro and ex vivo cell-based assays to ascertain its anti-inflammatory potential. The compound was evaluated in murine models of lipopolysaccharide-induced endotoxemia and pulmonary neutrophilia. Parameters of airway inflammation, airway hyperreactivity and bronchoconstriction were evaluated in a guinea pig model of antigen-induced allergy. In order to assess the emetic potential, the compound was evaluated biochemically for binding to high-affinity rolipram-binding site. Subsequently, the compound was tested in a surrogate model for emesis, and the results obtained were correlated directly to tests conducted in a Beagle dog model. RESULTS: NIS-62949 is a potent, highly selective PDE4 inhibitor. The compound demonstrated potent ability to inhibit tumor necrosis factor-alpha release from human peripheral blood mononuclear cells, lymphocyte proliferation and cytokine production. The in vitro profile of NIS-62949 prompted further evaluation of the compound in vivo and the compound was found to be comparable to roflumilast in several experimental models of pulmonary inflammation. Importantly, NIS-62949 displayed a safer profile compared to roflumilast. CONCLUSIONS: Our results report the development of a promising, novel PDE4 inhibitor, NIS-62949, with a wider therapeutic window as compared to second-generation PDE4 inhibitors such as roflumilast.


Subject(s)
Inflammation/drug therapy , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Animals , Carboxylic Acids/pharmacology , Cell Line, Transformed , Cyclic AMP/metabolism , Cyclohexanecarboxylic Acids , Disease Models, Animal , Dogs , Female , Guinea Pigs , Humans , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred BALB C , Nitriles/pharmacology , Rats , Rats, Wistar , Rolipram/metabolism , Rolipram/pharmacology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism , Vomiting/drug therapy
8.
Curr Opin Investig Drugs ; 8(5): 364-72, 2007 May.
Article in English | MEDLINE | ID: mdl-17520865

ABSTRACT

Intracellular levels of cyclic nuclec tides are closely regulated by distinct families of PD Es, which are responsible for the breakdown and degradation of cyclic nucleotides within cells. Type 4 PDEs have the potency to modulate the release of inflammatory mediators through cAMP-dependent and -independent mechanisms. Selective targeting of PDE4 is currently being investigated as a novel therapeutic approach in the treatment of inflammation-associated respiratory diseases such as asthma and COPD. The development of several PDE4 inhibitors, including roflumilast and cilomilast, reflects the success of this approach. In principle, therapeutic intervention of an inflammatory response by PDE4 inhibitors may be extended to other chronic inflammatory disease states such as psoriasis, rheumatoid arthritis and inflammatory bowel diseases (e.g., Crohns disease and ulcerative colitis). This retiiew explores the feasibility of PDE4 inhibitors as a promising alternative for therapeutic intervention in systemic inflammation and inflammation-based disease.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Anti-Inflammatory Agents/therapeutic use , Drugs, Investigational/therapeutic use , Inflammation/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/adverse effects , Arthritis/drug therapy , Asthma/drug therapy , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4 , Dermatitis, Atopic/drug therapy , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Humans , Inflammation/enzymology , Inflammatory Bowel Diseases/drug therapy , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/adverse effects , Psoriasis/drug therapy , Pulmonary Disease, Chronic Obstructive/drug therapy , Signal Transduction/drug effects
9.
PLoS One ; 10(2): e0118096, 2015.
Article in English | MEDLINE | ID: mdl-25692288

ABSTRACT

Induction of potent antibody is the goal of many vaccines targeted against infections or cancer. Modern vaccine designs that use virus-like particles (VLP) have shown efficacy for prophylactic vaccination against virus-associated cancer in the clinic. Here we used plant viral particles (PVP), which are structurally analogous to VLP, coupled to a weak idiotypic (Id) tumour antigen, as a conjugate vaccine to induce antibody against a murine B-cell malignancy. The Id-PVP vaccine incorporates a natural adjuvant, the viral ssRNA, which acts via TLR7. It induced potent protective anti-Id antibody responses in an in vivo mouse model, superior to the "gold standard" Id vaccine, with prevalence of the IgG2a isotype. Combination with alum further increased antibody levels and maintained the IgG2a bias. Engagement of TLR7 in vivo was followed by secretion of IFN-α by plasmacytoid dendritic cells and by activation of splenic CD11chi conventional dendritic cells. The latter was apparent from up-regulation of co-stimulatory molecules and from secretion of a wide range of inflammatory cytokines and chemokines including the Th1-governing cytokine IL-12, in keeping with the IgG2a antibody isotype distribution. PVP conjugates are a novel cancer vaccine design, offering an attractive molecular form, similar to VLP, and providing T-cell help. In contrast to VLP, they also incorporate a safe "in-built" ssRNA adjuvant.


Subject(s)
Antibodies, Viral/immunology , Plant Viruses/immunology , Toll-Like Receptor 7/metabolism , Animals , Dendritic Cells , Female , Immunoglobulin G/immunology , Interleukin-12/metabolism , Lymphoma , Male , Mice , Mice, Inbred C57BL , Nicotiana/virology , Toll-Like Receptor 7/genetics , Virion/immunology
10.
J Immunol Methods ; 272(1-2): 55-65, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12505712

ABSTRACT

Interferon-gamma (IFN-gamma) is an important immunomodulatory and pleiotropic cytokine produced, primarily, by activated T lymphocytes and natural killer (NK) cells. We have devised a nitric oxide (NO)-based bioassay for mouse IFN-gamma using resident peritoneal exudate cells (PECs) from C57BL/6 mice. Comparison with three existing bioassays demonstrated that this assay was very sensitive and detected IFN-gamma in the linear range of approximately 0.03-0.25 U/ml. Other cytokines, e.g. interleukin (IL)-2, IL-4, IL-6, IFN-alpha/beta and tumor necrosis factor-alpha (TNF-alpha), either alone or in combination with IFN-gamma, did not greatly modulate NO levels produced by resident peritoneal exudate cells. The presence of exogenous NO(3)(-) and H(2)O(2) did not interfere with the IFN-gamma induced NO production and detection. We also showed that the effect of lipopolysaccharide (LPS), which may be present in samples, could be suppressed by the use of Polymyxin B in the bioassay. The high sensitivity of the bioassay permitted the detection of low amounts of IFN-gamma in 1% mouse serum. In addition, this assay reproducibly detected bioactive IFN-gamma amounts in supernatants of activated T cells. The increase in IFN-gamma production by activated T cells in response to CD28 costimulation was approximately 3-fold by this bioassay and approximately 5-fold by ELISA. In summary, we have devised a simple, sensitive, inexpensive and high throughput method for the reproducible detection of bioactive IFN-gamma.


Subject(s)
Biological Assay/methods , Interferon-gamma/analysis , Nitric Oxide/biosynthesis , Animals , Ascitic Fluid/cytology , Ascitic Fluid/immunology , Ascitic Fluid/metabolism , Biological Assay/standards , Biological Assay/statistics & numerical data , Cell Line , Cytokines/pharmacology , Female , In Vitro Techniques , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Polymyxin B/pharmacology , Sensitivity and Specificity , T-Lymphocytes/immunology
11.
Indian J Med Res ; 120(2): 75-85, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15347856

ABSTRACT

The immune system needs to recognise target protein antigens from pathogens residing in both extracellular and intracellular locations. Intricate proteolytic processing events that follow antigen/ pathogen encounter provide the immune system with a complex display of a heterogeneous peptide mix, instrumental in the initiation of T cell immune responses, and allow the separation of extracellular and intracellular pathogen identification. However, recent evidence shows that this conventional dimorphism in the proteolytic processing of endogenous versus internalised antigen is less restrictive than originally recognized. The events that constitute the conventional major histocompatibility complex (MHC)-restricted processing pathways are accompanied by interesting deviations that provide novel adjuncts for the processing machinery to gain access to antigen in varied intracellular locations. This review discusses these aspects of classical and non-classical processing pathways for MHC-restricted protein presentation, which play significant roles in both optimising and diversifying the peptide repertoire available for immune recognition.


Subject(s)
Antigen Presentation , Major Histocompatibility Complex/immunology , T-Lymphocytes/immunology , Antigens/immunology , Antigens/metabolism , Endopeptidases/metabolism , Histocompatibility Antigens Class I , Histocompatibility Antigens Class II , Humans , Molecular Chaperones/metabolism
12.
Philos Trans R Soc Lond B Biol Sci ; 364(1517): 605-11, 2009 Mar 12.
Article in English | MEDLINE | ID: mdl-19008198

ABSTRACT

Somatic hypermutation of immunoglobulin (Ig) genes occurs at a frequency that is a million times greater than the mutation in other genes. Mutations occur in variable genes to increase antibody affinity, and in switch regions before constant genes to cause switching from IgM to IgG. Hypermutation is initiated in activated B cells when the activation-induced deaminase protein deaminates cytosine in DNA to uracil. Uracils can be processed by either a mutagenic pathway to produce mutations or a non-mutagenic pathway to remove mutations. In the mutagenic pathway, we first studied the role of mismatch repair proteins, MSH2, MSH3, MSH6, PMS2 and MLH1, since they would recognize mismatches. The MSH2-MSH6 heterodimer is involved in hypermutation by binding to U:G and other mismatches generated during repair synthesis, but the other proteins are not necessary. Second, we analysed the role of low-fidelity DNA polymerases eta, iota and theta in synthesizing mutations, and conclude that polymerase eta is the dominant participant by generating mutations at A:T base pairs. In the non-mutagenic pathway, we examined the role of the Cockayne syndrome B protein that interacts with other repair proteins. Mice deficient in this protein had normal hypermutation and class switch recombination, showing that it is not involved.


Subject(s)
Antibodies/genetics , Base Pair Mismatch/genetics , DNA-Directed DNA Polymerase/metabolism , MutS DNA Mismatch-Binding Protein/metabolism , Somatic Hypermutation, Immunoglobulin/genetics , Animals , Mice , Models, Genetic , Mutation/genetics , Somatic Hypermutation, Immunoglobulin/immunology , Uracil/metabolism
13.
J Exp Med ; 206(6): 1237-44, 2009 Jun 08.
Article in English | MEDLINE | ID: mdl-19433618

ABSTRACT

Repetitive DNA sequences in the immunoglobulin switch mu region form RNA-containing secondary structures and undergo hypermutation by activation-induced deaminase (AID). To examine how DNA structure affects transcription and hypermutation, we mapped the position of RNA polymerase II molecules and mutations across a 5-kb region spanning the intronic enhancer to the constant mu gene. For RNA polymerase II, the distribution was determined by nuclear run-on and chromatin immunoprecipitation assays in B cells from uracil-DNA glycosylase (UNG)-deficient mice stimulated ex vivo. RNA polymerases were found at a high density in DNA flanking both sides of a 1-kb repetitive sequence that forms the core of the switch region. The pileup of polymerases was similar in unstimulated and stimulated cells from Ung(-/-) and Aid(-/-)Ung(-/-) mice but was absent in cells from mice with a deletion of the switch region. For mutations, DNA was sequenced from Ung(-/-) B cells stimulated in vivo. Surprisingly, mutations of A nucleotides, which are incorporated by DNA polymerase eta, decreased 10-fold before the repetitive sequence, suggesting that the polymerase was less active in this region. We propose that altered DNA structure in the switch region pauses RNA polymerase II and limits access of DNA polymerase eta during hypermutation.


Subject(s)
Base Sequence , DNA , Immunoglobulin Switch Region/genetics , Nucleic Acid Conformation , RNA Polymerase II/genetics , Somatic Hypermutation, Immunoglobulin , Animals , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA/chemistry , DNA/genetics , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Transcription, Genetic , Uracil-DNA Glycosidase/genetics , Uracil-DNA Glycosidase/metabolism
14.
Eur J Immunol ; 36(4): 828-41, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16552710

ABSTRACT

The heat shock protein (HSP) Hsp90 is known to chaperone cytosolic peptides for MHC class I (MHCI)-restricted antigen presentation to T lymphocytes. We now demonstrate a role for Hsp90 activity in presentation of antigens on MHCII. Treatment of mouse antigen-presenting cells (APC) with the pharmacological Hsp90 inhibitor, geldanamycin, inhibited MHCII-mediated presentation of endocytosed and cytosolic proteins as well as synthetic peptides to specific T cells. Ectopic expression of human Hsp90 in APC enhanced MHCII-mediated antigen presentation. Further, pharmacological Hsp90 inhibition reduced, while retroviral Hsp90 overexpression enhanced, the levels of stable compact MHCII heterodimers correlating with the antigen presentation phenotype. Pharmacological inhibition of Hsp90 activity in IFN-gamma-treated APC resulted in severe abrogation of MHCII-restricted presentation of cytosolic antigen, but only partially inhibited exogenous antigen presentation. Our data suggest a major role for Hsp90 activity in MHCII-mediated antigen presentation pathways, and implicate IFN-gamma-inducible Hsp90-independent mechanisms.


Subject(s)
Antigen Presentation/immunology , HSP90 Heat-Shock Proteins/immunology , Histocompatibility Antigens Class II/immunology , Macrophages/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation/drug effects , Benzoquinones , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Flow Cytometry , HSP90 Heat-Shock Proteins/metabolism , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/metabolism , Humans , Interferon-gamma/immunology , Lactams, Macrocyclic , Lymphocyte Activation/immunology , Macrophages/drug effects , Mice , Quinones/pharmacology , T-Lymphocytes/drug effects , Transduction, Genetic
15.
Nat Immunol ; 6(3): 287-94, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15711549

ABSTRACT

By convention, presentation of major histocompatibility complex (MHC) class I-restricted epitopes involves processing by cytosolic proteasomes, whereas MHC class II-restricted epitopes are generated by endosomal proteases. Here, we show that two MHC class II-restricted epitopes within influenza virus were generated by a proteasome- and TAP-dependent pathway that was accessed by exogenous virus in dendritic cells (DCs) but not cell types with less permeable endosomes. Both epitopes were presented by recycling MHC class II molecules. Challenging mice with influenza or vaccinia viruses demonstrated that a substantial portion of the MHC class II-restricted response was directed against proteasome-dependent epitopes. By complementing endosomal activities, this pathway broadens the array of MHC class II-restricted epitopes available for CD4(+) T cell activation.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Antigen Presentation , Cytoplasm/metabolism , Histocompatibility Antigens Class II/metabolism , Proteasome Endopeptidase Complex/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , Animals , Female , Mice , Signal Transduction
16.
J Immunol ; 169(5): 2545-52, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12193724

ABSTRACT

Clearance of facultative intracellular pathogens such as Salmonella requires IFN-gamma from CD4 T cells. Mechanisms linking intracellular pathogen recognition with induction of IFN-gamma-producing T cells are still poorly understood. We show in this study that IL-12 is not required for commitment to the IFN-gamma-producing T cell response in infection with Salmonella typhimurium, but is needed for its maintenance. The IL-12-independent signals required for commitment depend on events during the first hour of infection and are related to Ag presentation. Even transient attenuation of Ag presentation early during infection specifically abrogates the IFN-gamma component of the resulting CD4 T cell response. The IL-12 needed for maintenance is also better induced by live rather than dead bacteria in vivo, and this difference is due to specific suppression of IL-12 induction by dead bacteria. Presence of exogenous IL-4 down-modulates IL-12 production by macrophages activated in vitro. Furthermore, macrophages from IL-4-null mice secrete high levels of both IL-12 and IL-18 in response to stimulation in vivo even with dead bacteria, but this does not lead to induction of IFN-gamma-secreting T cells in response to immunization with dead S. typhimurium. Early IL-4 is contributed by triggering of CD4 NK T cells by dead, but not live, bacteria. Thus, Ag presentation-related IL-12-independent events and IL-4-sensitive IL-12-dependent events play crucial complementary roles in the generation of the IFN-gamma-committed CD4 T cell component of the immune response in Salmonella infection.


Subject(s)
Interferon-gamma/biosynthesis , Interleukin-12/physiology , Salmonella typhimurium/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , Animals , Antigen Presentation/drug effects , Cells, Cultured , Chloroquine/administration & dosage , Histocompatibility Antigens Class I/physiology , Injections, Intraperitoneal , Interleukin-12/antagonists & inhibitors , Interleukin-12/biosynthesis , Interleukin-12/genetics , Interleukin-18/biosynthesis , Interleukin-4/deficiency , Interleukin-4/genetics , Interleukin-4/pharmacology , Killer Cells, Natural/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Mice , Mice, Knockout , Recombinant Proteins/pharmacology , Salmonella Infections, Animal/genetics , Salmonella Infections, Animal/immunology , Salmonella Vaccines/administration & dosage , Salmonella Vaccines/immunology , Salmonella typhimurium/drug effects , Salmonella typhimurium/growth & development , T-Lymphocyte Subsets/drug effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
17.
J Cell Sci ; 117(Pt 18): 4219-30, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15316082

ABSTRACT

Antigen-presenting cells (APCs) are expected to present peptides from endocytosed proteins via major histocompatibility complex (MHC) class II (MHCII) molecules to T cells. However, a large proportion of peptides purified from MHCII molecules are derived from cytosolic self-proteins making the pathway of cytosolic peptide loading onto MHCII of critical relevance in the regulation of immune self-tolerance. We show that peptides derived from cytoplasmic proteins either introduced or expressed in the cytoplasm are first detectable as MHCII-peptide complexes in LAMP-1(+) lysosomes, prior to their delivery to the cell surface. These peptide-MHC complexes are formed in a variety of APCs, including peritoneal macrophages, dendritic cells, and B cells, and are able to activate T cells. This process requires invariant chain (Ii)-dependent sorting of MHCII to the lysosome and the activity of the molecular chaperone H-2M. This pathway is independent of the ER resident peptide transporter complex TAP and does not take place by cross-presentation from neighbouring cells. In conjunction with our earlier results showing that these peptides are derived by cytosolic processing via the proteasome, these observations provide evidence for a ubiquitous route for peptide transport into the lysosome for the efficient presentation of endogenous and cytoplasmic proteins to CD4 T cells.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Endosomes/immunology , Histocompatibility Antigens Class II/immunology , Lysosomes/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/immunology , ATP-Binding Cassette Transporters/metabolism , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , Cell Line , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Endosomes/metabolism , Histocompatibility Antigens Class II/metabolism , Lysosomal Membrane Proteins , Lysosomes/metabolism , Macromolecular Substances/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/immunology , Molecular Chaperones/metabolism , Peptides/immunology , Peptides/metabolism , Protein Transport/physiology , Signal Transduction/immunology
18.
J Immunol ; 170(3): 1362-73, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12538696

ABSTRACT

Retro-inverso (ri) analogs of model T cell and B cell epitopes were predictively designed as mimics and then assayed for activity to understand the basis of functional ri-antigenic peptide mimicry. ri versions of two MHC class I binding peptide epitopes, one from a vesicular stomatitis virus glycoprotein (VSV(p)) and another from OVA (OVAp), exhibit structural as well as functional mimicry of their native counterparts. The two ri peptides exhibit conformational plasticity and they bind to MHC class I (H-2K(b)) similar to their native counterparts both in silico and in vivo. In fact, ri-OVAp is also presented to an OVAp-specific T cell line in a mode similar to native OVAp. In contrast, the ri version of an immunodominant B cell peptide epitope from a hepatitis B virus protein, PS1, exhibits no structural or functional correlation with its native counterpart. PS1 and its ri analog do not exhibit similar conformational propensities. PS1 is less flexible relative to its ri version. These observed structure-function relationships of the ri-peptide epitopes are consistent with the differences in recognition properties between peptide-MHC vs peptide-Ab binding where, while the recognition of the epitope by MHC is pattern based, the exquisitely specific recognition of Ag by Ab arises from the high complementarity between the Ag and the binding site of the Ab. It is evident that the correlation of conformational and interaction propensities of native L-peptides and their ri counterparts depends both on their inherent structural properties and on their mode of recognition.


Subject(s)
Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/metabolism , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/metabolism , Molecular Mimicry/immunology , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Binding Sites, Antibody , Cell Line , Egg Proteins/chemical synthesis , Egg Proteins/immunology , Egg Proteins/metabolism , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , H-2 Antigens/chemistry , H-2 Antigens/immunology , H-2 Antigens/metabolism , Hepatitis B Surface Antigens/chemistry , Hepatitis B Surface Antigens/immunology , Hepatitis B Surface Antigens/metabolism , Hydrophobic and Hydrophilic Interactions , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Ovalbumin/chemical synthesis , Ovalbumin/immunology , Ovalbumin/metabolism , Peptide Fragments/immunology , Protein Binding/immunology , Protein Conformation , Vesicular stomatitis Indiana virus/chemistry , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL