Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Acoust Soc Am ; 146(1): EL1, 2019 07.
Article in English | MEDLINE | ID: mdl-31370609

ABSTRACT

An unsupervised automatic clustering algorithm (k-means) classified 1282 Mel frequency cepstral coefficient (MFCC) representations of isolated steady-state vowel utterances from eight standard German vowel categories with fo between 196 and 698 Hz. Experiment I obtained the number of MFCCs (1-20) in connection with the spectral bandwidth (2-20 kHz) at which performance peaked (five MFCCs at 4 kHz). In experiment II, classification performance with different ranges of fo revealed that ranges with fo > 500 Hz reduced classification performance but it remained well above chance. This shows that isolated steady state vowels with strongly undersampled spectra contain sufficient acoustic information to be classified automatically.

2.
J Acoust Soc Am ; 142(2): 1025, 2017 08.
Article in English | MEDLINE | ID: mdl-28863619

ABSTRACT

The phonological function of vowels can be maintained at fundamental frequencies (fo) up to 880 Hz [Friedrichs, Maurer, and Dellwo (2015). J. Acoust. Soc. Am. 138, EL36-EL42]. Here, the influence of talker variability and multiple response options on vowel recognition at high fos is assessed. The stimuli (n = 264) consisted of eight isolated vowels (/i y e ø ε a o u/) produced by three female native German talkers at 11 fos within a range of 220-1046 Hz. In a closed-set identification task, 21 listeners were presented excised 700-ms vowel nuclei with quasi-flat fo contours and resonance trajectories. The results show that listeners can identify the point vowels /i a u/ at fos up to almost 1 kHz, with a significant decrease for the vowels /y ε/ and a drop to chance level for the vowels /e ø o/ toward the upper fos. Auditory excitation patterns reveal highly differentiable representations for /i a u/ that can be used as landmarks for vowel category perception at high fos. These results suggest that theories of vowel perception based on overall spectral shape will provide a fuller account of vowel perception than those based solely on formant frequency patterns.

3.
J Acoust Soc Am ; 138(1): EL36-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26233058

ABSTRACT

In a between-subject perception task, listeners either identified full words or vowels isolated from these words at F0s between 220 and 880 Hz. They received two written words as response options (minimal pair with the stimulus vowel in contrastive position). Listeners' sensitivity (A') was extremely high in both conditions at all F0s, showing that the phonological function of vowels can also be maintained at high F0s. This indicates that vowel sounds may carry strong acoustic cues departing from common formant frequencies at high F0s and that listeners do not rely on consonantal context phenomena for their identification performance.


Subject(s)
Phonetics , Speech Acoustics , Speech Perception/physiology , Adult , Analysis of Variance , Cues , Female , Humans , Male
4.
J Immunol ; 187(1): 164-71, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21622859

ABSTRACT

The IgE-mediated and Th2-dependent late-phase reaction remains a mechanistically enigmatic and daunting element of human allergic inflammation. In this study, we uncover the FcεRI on dendritic cells (DCs) as a key in vivo component of this form of allergy. Because rodent, unlike human, DCs lack FcεRI, this mechanism could be revealed only by using a new transgenic mouse model with human-like FcεRI expression on DCs. In the presence of IgE and allergen, FcεRI(+) DCs instructed naive T cells to differentiate into Th2 cells in vitro and boosted allergen-specific Th2 responses and Th2-dependent eosinophilia at the site of allergen exposure in vivo. Thus, FcεRI on DCs drives the cascade of pathogenic reactions linking the initial allergen capture by IgE with subsequent Th2-dominated T cell responses and the development of late-phase allergic tissue inflammation.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/pathology , Inflammation Mediators/metabolism , Receptors, IgE/metabolism , Th2 Cells/immunology , Th2 Cells/pathology , Allergens/toxicity , Animals , Antigens, Plant/toxicity , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Female , Humans , Inflammation Mediators/physiology , Inflammation Mediators/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/toxicity , Protein Binding/genetics , Protein Binding/immunology , Receptors, IgE/deficiency , Receptors, IgE/physiology , Th2 Cells/metabolism , Time Factors
5.
J Dtsch Dermatol Ges ; 8(12): 990-8, 2010 Dec.
Article in English, German | MEDLINE | ID: mdl-20678148

ABSTRACT

BACKGROUND: Our understanding of the pathogenic role of IgE in atopic dermatitis is incomplete. We asked whether blocking free IgE would alter the course of the disease. PATIENTS AND METHODS: We administered either omalizumab, a humanized monoclonal mouse antibody against IgE, or placebo subcutaneously for 16 weeks to 20 atopic dermatitis patients and measured immunological and clinical disease parameters. RESULTS: Omalizumab (I) reduced free serum IgE, (II) lowered surface IgE and FcɛRI expression on different peripheral blood mononuclear cells, (III) reduced the saturation of FcɛRI with IgE, (IV) increased the number of free FcɛRI and (V) lowered the number of IgE+, but not of FcɛRI+ cells in skin. The in vivo relevance of these results is evidenced by the increase in the threshold allergen concentration required to give a type I hypersensitivity reaction in the titrated skin test. While not significantly altering the clinical disease parameters, omalizumab treatment led to an improvement of the atopy patch test results in single patients, i.e. an eczematous reaction upon epicutaneous allergen challenge. CONCLUSIONS: The interference with immediate and delayed type skin tests may imply that a therapeutic benefit of omalizumab treatment, if present at all, would be seen in patients with acute rather than chronic forms of the disease.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Immunoglobulin E/immunology , Adolescent , Adult , Anti-Allergic Agents/therapeutic use , Antibodies, Anti-Idiotypic , Antibodies, Monoclonal, Humanized , Double-Blind Method , Female , Humans , Injections, Subcutaneous , Male , Middle Aged , Omalizumab , Pilot Projects , Placebo Effect , Skin Tests , Treatment Outcome , Young Adult
6.
J Immunol ; 183(2): 1133-43, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19570825

ABSTRACT

Depending on the cellular and molecular microenvironment, immune responses generated by skin-associated lymphoid tissues can lead to protective immunity against pathogens or to tolerance. In this study, we investigated immune responses to an Ag expressed de novo in adult skin under homeostatic conditions by generating transgenic mice producing the Ag Ova in a Cre-inducible manner in keratinocytes. Expression of Ova was induced in adult mice with a tamoxifen-inducible K5-CreER transgenic line. Although Ova was efficiently expressed by keratinocytes and presented by Langerhans cells after Cre-mediated transgene recombination, adult transgenic mice did not develop any obvious autoimmune disease symptoms like hair or weight loss. Ag-specific T cells were activated after Ova expression as indicated by up-regulation of CD44 and CD69. After in vitro restimulation Ova-specific T cells showed reduced IFN-gamma production suggesting induction of tolerance after Ova expression in the skin. After transfer into Ova-expressing mice, naive OT-1 T cells transiently proliferated in skin-draining lymph nodes, infiltrated the skin but did not cause disease. Topical application of danger signals at the time of Ova induction did also not induce autoimmune disease. The unresponsiveness of Ag-specific T cells after induction of Ova expression could only be circumvented by simultaneous priming with CpG-matured, bone marrow-derived dendritic cells. Our data suggest that low amount of Ag expressed in the induction phase of the immune response results in tolerance even in the presence of danger signals and thereby helps to preserve homeostasis in the skin under normal and pathologic conditions.


Subject(s)
Immune Tolerance , Inflammation/immunology , Isoantigens/immunology , Skin/pathology , Animals , Dendritic Cells/immunology , Isoantigens/genetics , Keratinocytes/metabolism , Langerhans Cells/immunology , Mice , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
7.
J Immunol ; 179(6): 4272-82, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785868

ABSTRACT

Viruses can escape destruction by the immune system by exploitation of the chemokine-chemokine receptor system. It is less established whether human cancers can adopt similar strategies to evade immunologic control. In this study, we show that advanced cutaneous T cell lymphoma (CTCL) is associated with selective and efficient inactivation of CXCR3-dependent T cell migration. Our studies demonstrate that this alteration is at least in part due to CXCR3 down-regulation in vivo by elevated serum levels of CXCR3 ligands. The T cell population most affected by this down-regulatory mechanism are CD8+ cytotoxic effector T cells. In CTCL patients, cytotoxic effector T cells have strongly reduced surface CXCR3 expression, accumulate in peripheral blood, but are virtually absent from CTCL tumor lesions, indicating an inability to extravasate into lymphoma tissue. CTCL-associated inactivation of effector cell recruitment may be a paradigmatic example of a new type of immune escape mechanisms shielding the neoplasm from a tumoricidal attack.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Down-Regulation/immunology , Lymphoma, T-Cell, Cutaneous/immunology , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/biosynthesis , Skin Neoplasms/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Membrane , Cell Movement/immunology , Cells, Cultured , E-Selectin/biosynthesis , E-Selectin/metabolism , Endosomes/metabolism , Endothelial Cells/metabolism , Humans , Immunologic Memory , K562 Cells , L-Selectin/biosynthesis , Ligands , Lymphoma, T-Cell, Cutaneous/metabolism , Lymphoma, T-Cell, Cutaneous/pathology , Lymphoma, T-Cell, Cutaneous/therapy , Lysosomes/metabolism , Receptors, CXCR3 , Receptors, Chemokine/physiology , Resting Phase, Cell Cycle/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Tumor Cells, Cultured , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/metabolism
8.
J Immunol ; 178(4): 2262-71, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277131

ABSTRACT

It is unclear whether peptide-MHC class II (pMHC) complexes on distinct types of APCs differ in their capacity to trigger TCRs. In this study, we show that individual cognate pMHC complexes displayed by dendritic cells (DCs), as compared with nonprofessional APCs, are far better in productively triggering Ag-specific TCRs independently of conventional costimulation. As we further show, this is accomplished by the unique ability of DCs to robustly activate the Src family kinases (SFKs) Lck and Fyn in T cells even in the absence of cognate peptide. Instead, this form of SFK activation depends on interactions of DC-displayed MHC with TCRs of appropriate restriction, suggesting a central role of self-pMHC recognition. DC-mediated SFK activation leads to "TCR licensing," a process that dramatically increases sensitivity and magnitude of the TCR response to cognate pMHC. Thus, TCR licensing, besides costimulation, is a main mechanism of DCs to present Ag effectively.


Subject(s)
Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Proto-Oncogene Proteins c-fyn/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Antigen Presentation/immunology , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Autoimmunity , Cell Line, Transformed , Dendritic Cells/enzymology , Enzyme Activation/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Peptides/immunology , Peptides/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
9.
Blood ; 109(11): 4777-85, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17289814

ABSTRACT

The discovery of marker proteins of human blood (BECs) and lymphatic endothelial cells (LECs) has allowed researchers to isolate these cells. So far, efforts to unravel their transcriptional and functional programs made use of cultured cells only. Hence, it is unknown to which extent previously identified LEC- and BEC-specific programs are representative of the in vivo situation. Here, we define the human BEC- and LEC-specific in vivo transcriptomes by comparative genomewide expression profiling of freshly isolated cutaneous EC subsets and of non-EC skin cells (fibroblasts, mast cells, dendritic cells, epithelial cells). Interestingly, the expression of most of the newly identified EC subset-discriminating genes depends strictly on the in vivo tissue environment as revealed by comparative analyses of freshly isolated and cultured EC subsets. The identified environment-dependent, EC subset-restricted gene expression regulates lineage fidelity, fluid exchange, and MHC class II-dependent antigen presentation. As an example for a BEC-restricted in vivo function, we show that non-activated BECs in situ, but not in vitro, assemble and display MHC class II protein complexes loaded with self-peptides. Thus, our data demonstrate the key importance of using precisely defined native ECs for the global identification of in vivo relevant cell functions.


Subject(s)
Endothelial Cells/cytology , Gene Expression Profiling , Melanoma/immunology , Cell Differentiation , Cell Separation , Cells, Cultured/metabolism , Genome , Histocompatibility Antigens Class II/chemistry , Humans , Microcirculation , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Peptides/chemistry , Transcription, Genetic
10.
FEBS J ; 274(7): 1747-58, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17313482

ABSTRACT

Prion diseases are fatal neurodegenerative disorders caused by proteinaceous infectious pathogens termed prions (PrP(Sc)). To date, there is no prophylaxis or therapy available for these transmissible encephalopathies. Passive immunization with monclonal antibodies recognizing the normal host-encoded prion protein (PrP(C)) has been reported to abolish PrP(Sc) infectivity and to delay onset of disease. Because of established immunologic tolerance against the widely expressed PrP(C), active immunization appears to be difficult to achieve. To overcome this limitation, papillomavirus-like particles were generated that display a nine amino acid B-cell epitope, DWEDRYYRE, of the murine/rat prion protein in an immunogenic capsid surface loop, by insertion into the L1 major capsid protein of bovine papillomavirus type 1. The PrP peptide was selected on the basis of its previously suggested central role in prion pathogenesis. Immunization with PrP-virus-like particles induced high-titer antibodies to PrP in rabbit and in rat, without inducing overt adverse effects. As determined by peptide-specific ELISA, rabbit immune sera recognized the inserted murine/rat epitope and also cross-reacted with the homologous rabbit/human epitope differing in one amino acid residue. In contrast, rat immune sera recognized the murine/rat peptide only. Sera of both species reacted with PrP(C) in its native conformation in mouse brain and on rat pheochromocytoma cells, as determined by immunoprecipitation and fluorescence-activated cell sorting analysis. Importantly, rabbit anti-PrP serum contained high-affinity antibody that inhibited de novo synthesis of PrP(Sc) in prion-infected cells. If also effective in vivo, PrP-virus-like particle vaccination opens a unique possibility for immunologic prevention of currently fatal and incurable prion-mediated diseases.


Subject(s)
Bovine papillomavirus 1/immunology , Peptide Fragments/immunology , PrPC Proteins/immunology , PrPSc Proteins/immunology , Prion Diseases/prevention & control , Vaccination/methods , Amino Acid Sequence , Animals , Antibody Affinity/immunology , Bovine papillomavirus 1/genetics , Capsid Proteins/genetics , Capsid Proteins/immunology , Cell Line, Tumor , Epitopes/genetics , Epitopes/immunology , Flow Cytometry , Haptens/genetics , Haptens/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , PC12 Cells , Peptide Fragments/genetics , PrPC Proteins/genetics , Prion Diseases/immunology , Rabbits , Rats , Rats, Inbred Lew , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/ultrastructure
11.
Blood ; 109(10): 4288-95, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17255360

ABSTRACT

Liver X receptors (LXRs) are nuclear receptors regulating lipid and cholesterol metabolism. Recent data revealed a cross talk between LXR and Toll-like receptor signaling in macrophages, indicating a role in immunity. Here, we show that LXRalpha is expressed in human myeloid dendritic cells (DCs) and induced during differentiation of monocyte-derived DCs, whereas LXRbeta is expressed constitutively at a very low level. LXR activation by 2 different LXR agonists strongly interfered with lipopolysaccharide (LPS)-induced but not with CD40L-induced DC maturation by altering DC morphology and suppressing interleukin-12-but enhancing interleukin-10-secretion. LXR activation in DCs largely blocked their T-cell stimulatory ability despite essentially unaltered expression of various antigen-presenting and costimulatory molecules. Immunologic synapse formation was significantly inhibited by LXR activation along with a complete block in LPS- but not CD40L-induced expression of the actin-bundling protein fascin. Notably, overexpression of fascin in LXR agonist-treated DCs restored immunologic synapse formation and restored their ability to activate T cells. In conclusion, our data reveal LXR as a potent modulator of DC maturation and function mediated in part by blocking the expression of fascin. Due to the central position of DCs in immunity, LXRalpha could be a potential novel target for immunomodulation.


Subject(s)
Carrier Proteins/genetics , DNA-Binding Proteins/physiology , Dendritic Cells/cytology , Dendritic Cells/physiology , Microfilament Proteins/genetics , Phenotype , Receptors, Cytoplasmic and Nuclear/physiology , Carrier Proteins/metabolism , Cells, Cultured , DNA-Binding Proteins/agonists , DNA-Binding Proteins/metabolism , Dendritic Cells/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Hydrocarbons, Fluorinated , Liver X Receptors , Male , Microfilament Proteins/metabolism , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Sulfonamides/pharmacology , Transfection
12.
Blood ; 106(1): 175-83, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15755895

ABSTRACT

The life cycle of dendritic cells (DCs) must be precisely regulated for proper functioning of adaptive immunity. However, signaling pathways actively mediating DC death remain enigmatic. Here we describe a novel mechanism of hierarchical transcriptional control of DC life and death. Ligation of tumor necrosis factor receptor superfamily (TNFR-SF) members on DCs and cognate contact with T cells resulted in quantitatively balanced nuclear factor-kappaB (NF-kappaB) and c-Jun N-terminal kinase (JNK)-mediated activator protein-1 (AP-1) induction and strongly enhanced DC longevity. Specific blockade of NF-kappaB in DCs induced strongly augmented JNK/AP-1 activity because of elevated levels of reactive oxygen species. In this scenario, DC activation by TNFR-SF members or T cells induced DC apoptosis. Specific inhibition of JNK/AP-1 rescued DCs from this activation-induced cell death program and restored TNFR-SF member- and T-cell-mediated survival. We conclude that JNK/AP-1 activity is under negative feedback control of NF-kappaB and can execute apoptosis in DCs. Thus, feedback-controlled signaling amplitudes of 2 transcriptional pathways decide the fate of a DC.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , CD40 Antigens/metabolism , Cell Communication/immunology , Cell Death/immunology , Cells, Cultured , Coculture Techniques , Epidermal Cells , Humans , I-kappa B Proteins/genetics , Langerhans Cells/cytology , Langerhans Cells/metabolism , Mitochondria/metabolism , NF-KappaB Inhibitor alpha , Oxidative Stress/immunology , Signal Transduction/immunology , T-Lymphocytes/cytology , Transcriptional Activation/immunology , Transfection
13.
J Immunol ; 173(11): 6592-602, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15557149

ABSTRACT

Plasmacytoid dendritic cells (pDCs) recognize microbes, viruses in particular, and provide unique means of innate defense against them. The mechanism of pDC tissue recruitment remained enigmatic because the ligands of CXCR3, the cardinal chemokine receptor on pDCs, have failed to induce in vitro chemotaxis of pDCs in the absence of additional chemokines. In this study, we demonstrate that CXCR3 is sufficient to induce pDC migration, however, by a migratory mechanism that amalgamates the features of haptotaxis and chemorepulsion. To mediate "haptorepulsion" of pDCs, CXCR3 requires the encounter of its cognate ligands immobilized, optimally by heparan sulfate, in a form of a negative gradient. This is the first report of the absolute requirement of chemokine immobilization and presentation for its in vitro promigratory activity. The paradigmatic example of pDC haptorepulsion described here may represent a new pathophysiologically relevant migratory mechanism potentially used by other cells in response to other chemokines.


Subject(s)
Cell Movement/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Receptors, Chemokine/physiology , Antigen Presentation , Cell Adhesion/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Migration Inhibition , Chemokine CXCL10 , Chemokine CXCL12 , Chemokines/pharmacology , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Chemokines, CXC/physiology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Heparitin Sulfate/immunology , Heparitin Sulfate/metabolism , Herpes Zoster/immunology , Herpes Zoster/pathology , Herpesvirus 3, Human/immunology , Humans , Interferon-alpha/pharmacology , Ligands , Pertussis Toxin/pharmacology , Phosphorylation , Protein Binding/immunology , Receptors, CCR7 , Receptors, CXCR3 , Receptors, Chemokine/biosynthesis , Simplexvirus/immunology , Solubility , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tyrosine/metabolism
14.
J Immunol ; 173(3): 1628-39, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15265891

ABSTRACT

Their eponymous morphology and unique ability to activate naive T cells are hallmark features of dendritic cells (DCs). Specific properties of the actin cytoskeleton may define both characteristics. In search for regulators that coordinate DC phenotype and function, we observed strongly increased expression of the actin-remodeling GTPases Cdc42 and Rac1 during DC development from human stem cells. Cdc42 and Rac1 are constitutively active in immature DCs, and their activity is further up-regulated by maturational stimuli such as LPS or CD40L. Activation of Rac1 is associated with its rapid recruitment into lipid rafts. Cdc42 is not recruited into rafts, but readily activated by raft-associated moieties. The functional interplay of rafts, GTPases, and cortical actin is further shown by GTPase activation and actin remodeling after pharmacological disruption of lipid rafts and by the loss of the actin-based DC morphology by transfection of dominant-negative Cdc42 and Rac1. Both Cdc42 and Rac1 also control the transport of essential immunostimulatory molecules to the DC surface. Transfection with dominant-negative GTPases led to reduced surface expression of MHC class I and CD86. Consecutively, DCs display a reduced stimulatory capacity for CD8(+) T cells, whereas MHC class II-dependent stimulation of CD4(+) T cells remains unperturbed. We conclude that Cdc42 and Rac1 signaling controls DC morphology and conditions DCs for efficient CD8(+) T cell stimulation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/enzymology , Lymphocyte Activation/physiology , Membrane Microdomains/enzymology , Neuropeptides/physiology , beta-Cyclodextrins , cdc42 GTP-Binding Protein/physiology , rac GTP-Binding Proteins , rac1 GTP-Binding Protein/physiology , Actins/metabolism , Antigens, CD/metabolism , B7-2 Antigen , Biological Transport/physiology , CD40 Ligand/pharmacology , Cyclodextrins/pharmacology , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Dendritic Cells/immunology , Dendritic Cells/ultrastructure , Enzyme Activation , Enzyme Induction , Genes, Dominant , Histocompatibility Antigens Class I/metabolism , Humans , Interleukin-12/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Culture Test, Mixed , Macrophages/ultrastructure , Membrane Glycoproteins/metabolism , Membrane Microdomains/drug effects , Neuropeptides/deficiency , Neuropeptides/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transfection , cdc42 GTP-Binding Protein/deficiency , cdc42 GTP-Binding Protein/genetics , p21-Activated Kinases , rac1 GTP-Binding Protein/deficiency , rac1 GTP-Binding Protein/genetics
15.
J Immunol ; 171(5): 2714-24, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12928425

ABSTRACT

Therapeutic vaccination against cutaneous T cell lymphoma (CTCL) requires the characterization of cancer cell-specific CTL epitopes. Despite reported evidence for tumor-reactive cytotoxicity in CTCL patients, the nature of the recognized determinants remains elusive. The clonotypic TCR of CTCL cells is a promising candidate tumor-specific Ag. In this study, we report that the clonotypic and framework regions of the TCRs expressed in the malignant T cell clones of six CTCL patients contain multiple peptides with anchor residues fitting the patients' MHC class I molecules. We demonstrate that TCR peptide-specific T cells from the blood of healthy donors and patients can be induced to become cytotoxic effectors after repeated stimulation with 6 of 11 selected peptides with experimentally proven affinity for HLA-A*0201. Importantly, 4 of these 6 CTL lines reproducibly recognize and lyse autologous primary CTCL cells in MHC class I/CD8-dependent fashion. These tumoricidal CTL lines are directed against epitopes from V, hypervariable, and C regions of TCRalpha. We therefore conclude that recombined as well as V framework regions of the tumor cell TCRs contain predictable epitopes for CTL-mediated attack of CTCL cells. Our data further suggest that such peptides represent valuable tools for future anti-CTCL vaccination approaches.


Subject(s)
Cancer Vaccines/immunology , Cytotoxicity, Immunologic/immunology , Epitopes, T-Lymphocyte/therapeutic use , Lymphoma, T-Cell/immunology , Receptors, Antigen, T-Cell/therapeutic use , Skin Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Antigen Presentation/immunology , Cancer Vaccines/metabolism , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Clone Cells , Endopeptidases/metabolism , Epitopes, T-Lymphocyte/blood , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Hydrolysis , Lymphocyte Activation , Lymphoma, T-Cell/blood , Lymphoma, T-Cell/enzymology , Lymphoma, T-Cell/prevention & control , Molecular Sequence Data , Peptide Fragments/blood , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding/immunology , Receptors, Antigen, T-Cell/blood , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Skin Neoplasms/blood , Skin Neoplasms/enzymology , Skin Neoplasms/prevention & control , T-Lymphocytes, Cytotoxic/enzymology , T-Lymphocytes, Cytotoxic/metabolism
16.
J Allergy Clin Immunol ; 110(3): 492-9, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12209101

ABSTRACT

BACKGROUND: Circulating autoantibodies against FcepsilonRI, IgE, or both occur in approximately one third of patients with chronic idiopathic urticaria (CIU), but not all autoantibodies initiate histamine release. OBJECTIVE: We sought to classify patients with CIU into subsets on the basis of serum bioactivity and immunoreactivity and to examine the relationship between newly defined subtype and disease severity. METHODS: Sera from patients with CIU (n = 78), dermog-raphism (n = 15), and cholinergic urticaria (n = 10) and sera from healthy subjects (n = 39) were analyzed by means of Western blot analysis for anti-FcepsilonRI autoantibodies and for histamine release from basophils and dermal mast cells. In vivo reactivity of autologous serum was tested by means of intradermal injection, and CIU severity was determined on the basis of clinical interview. RESULTS: We classified sera from patients with CIU into 5 subsets: immunoreactive histamine-releasing anti-FcepsilonRI autoantibodies (n = 20 [26%]); immunoreactive anti-FcepsilonRI autoantibodies without histamine-releasing activity (n = 12 [15%]); anti-IgE-like autoantibodies (n = 7 [9%]); serum containing a mast cell-specific histamine-releasing factor (n = 7 [9%]); and sera with no identifiable factor (n = 32 [41%]). Patients with serum histamine-releasing activity had more severe urticaria than patients without such activity. Positive skin test responses to autologous sera were associated with histamine-releasing anti-FcepsilonRI autoantibodies but not with non-histamine-releasing anti-FcepsilonRI autoantibodies. Neither healthy subjects nor patients with dermographism or cholinergic urticaria had his-tamine-releasing anti-FcepsilonRI autoantibodies. CONCLUSION: These data support the specificity of functional anti-FcepsilonRI autoantibodies to CIU. The identification of distinctive subsets of patients suggests that other pathogenic mechanisms occur in CIU in addition to direct ligation of FcepsilonRI by autoantibodies causing dermal mast cell degranulation. Elucidating these mechanisms might lead to new treatments for CIU.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Autoantibodies/immunology , Receptors, IgE/immunology , Urticaria/classification , Urticaria/diagnosis , Adolescent , Adult , Autoantibodies/blood , Blotting, Western , Chronic Disease , Histamine Release , Humans , Middle Aged , Skin Tests , Urticaria/immunology
SELECTION OF CITATIONS
SEARCH DETAIL