Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Parasite Immunol ; 37(2): 97-104, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25559085

ABSTRACT

Schistosome infections are renowned for their ability to induce regulatory networks such as regulatory T cells (Treg) that control immune responses against homologous and heterologous antigens such as allergies. However, in the case of co-infections with hepatitis C virus (HCV), schistosomes accentuate disease progression and we hypothesized that expanding schistosome-induced Treg populations change their phenotype and could thereby suppress beneficial anti-HCV responses. We therefore analysed effector T cells and n/iTreg subsets applying the markers Granzyme B (GrzB) and Helios in Egyptian cohorts of HCV mono-infected (HCV), schistosome-co-infected (Sm/HCV) and infection-free individuals. Interestingly, viral load and liver transaminases were significantly elevated in Sm/HCV individuals when compared to HCV patients. Moreover, overall Treg frequencies and Helios(pos) Treg were not elevated in Sm/HCV individuals, but frequencies of GrzB(+) Treg were significantly increased. Simultaneously, GrzB(+) CD8(+) T cells were not suppressed in co-infected individuals. This study demonstrates that in Sm/HCV co-infected cohorts, liver disease is aggravated with enhanced virus replication and Treg do not expand but rather change their phenotype with GrzB possibly being a more reliable marker than Helios for iTreg. Therefore, curing concurrent schistosome disease could be an important prerequisite for successful HCV treatment as co-infected individuals respond poorly to interferon therapy.


Subject(s)
Coinfection/immunology , Hepacivirus/physiology , Hepatitis C, Chronic/immunology , Schistosoma mansoni/physiology , Schistosomiasis mansoni/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Animals , Female , Humans , Interleukin-8/immunology , Liver/pathology , Liver/virology , Male , Middle Aged , Viral Load
2.
Clin Exp Immunol ; 178(2): 212-23, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24943899

ABSTRACT

Overall asthmatic symptoms can be controlled with diverse therapeutic agents. However, certain symptomatic individuals remain at risk for serious morbidity and mortality, which prompts the identification of novel therapeutic targets and treatment strategies. Thus, using an adjuvant-free T helper type 2 (Th2) murine model, we have deciphered the role of interleukin (IL)-1 signalling during allergic airway inflammation (AAI). Because functional IL-1ß depends on inflammasome activation we first studied asthmatic manifestations in specific inflammasome-deficient [NACHT, LRR and PYD domains-containing protein 3 (NLRP3(-/-) ) and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC(-/-) )] and IL-1 receptor type 1(-/-) (IL-1R1(-/-) ) mice on the BALB/c background. To verify the onset of disease we assessed cellular infiltration in the bronchial regions, lung pathology, airway hyperresponsiveness and ovalbumin (OVA)-specific immune responses. In the absence of NLRP3 inflammasome-mediated IL-1ß release all symptoms of AAI were reduced, except OVA-specific immunoglobulin levels. To address whether manipulating IL-1 signalling reduced asthmatic development, we administered the IL-1R antagonist anakinra (Kineret®) during critical immunological time-points: sensitization or challenge. Amelioration of asthmatic symptoms was only observed when anakinra was administered during OVA challenge. Our findings indicate that blocking IL-1 signalling could be a potential complementary therapy for allergic airway inflammation.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Respiratory Hypersensitivity/metabolism , Acute Disease , Animals , Apoptosis Regulatory Proteins/deficiency , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Cytokines/metabolism , Disease Models, Animal , Eosinophilia/genetics , Eosinophilia/immunology , Female , Goblet Cells/pathology , Hyperplasia , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Ovalbumin/adverse effects , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Receptors, Interleukin-1 Type I/deficiency , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
3.
Br J Cancer ; 104(6): 948-56, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21407224

ABSTRACT

BACKGROUND: The development of a successful immunotherapy is hampered by an ineffective T-cell repertoire against tumour antigens and the inability of the patient's immune system to overcome tolerance-inducing mechanisms. Here, we test the specific recognition and lytical potential of allo-restricted CD8(+) T cells against Ewing tumour (ET) associated antigens Enhancer of Zeste, Drosophila Homolog 2 (EZH2), and Chondromodulin-I (CHM1) identified through previous microarray analysis. METHODS: Following repetitive CHM1(319) (VIMPCSWWV) and EZH2(666) (YMCSFLFNL) peptide-driven stimulations with HLA-A 0201(+) dendritic cells (DC), allo-restricted HLA-A 0201(-) CD8(+) T cells were stained with HLA-A 0201/peptide multimers, sorted and expanded by limiting dilution. RESULTS: Expanded T cells specifically recognised peptide-pulsed target cells or antigen-transfected cells in the context of HLA-A 0201 and killed HLA-A 0201(+) ET lines expressing the antigen while HLA-A 0201(-) ET lines were not affected. Furthermore, adoptively transferred T cells caused significant ET growth delay in Rag2(-/-)γ(C)(-/-) mice. Within this context, we identified the CHM1(319) peptide as a new candidate target antigen for ET immunotherapy. CONCLUSION: These results clearly identify the ET-derived antigens, EZH2(666) and CHM1(319), as suitable targets for protective allo-restricted human CD8(+) T-cell responses against non-immunogenic ET and may benefit new therapeutic strategies in ET patients treated with allogeneic stem cell transplantation.


Subject(s)
Antigens, Neoplasm/immunology , Bone Neoplasms/pathology , Isoantigens/immunology , Sarcoma, Ewing/pathology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/metabolism , Bone Neoplasms/immunology , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Cytotoxicity, Immunologic/immunology , DNA-Binding Proteins/genetics , Down-Regulation , Humans , Immunotherapy, Adoptive , Isoantigens/metabolism , K562 Cells , Mice , Mice, Knockout , Sarcoma, Ewing/immunology , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocytes, Cytotoxic/metabolism
4.
J Exp Med ; 188(1): 61-70, 1998 Jul 06.
Article in English | MEDLINE | ID: mdl-9653084

ABSTRACT

The mechanisms underlying the genesis and maintenance of T cell memory remain unclear. In this study, we examined the evolution of a complex, antigen-specific T cell population during the transition from primary effector to memory T cells after Listeria monocytogenes infection. T cell populations specific for listeriolysin O (LLO)91-99, the immunodominant epitope recognized by H2-Kd-restricted T lymphocytes, were directly identified in immune spleens using tetrameric H2-Kd-epitope complexes. The T cell receptor (TCR) Vbeta repertoire of specific T cells was determined by direct, ex vivo staining with a panel of mAbs. We demonstrate that LLO91-99-specific, primary effector T cell populations have a diverse TCR Vbeta repertoire. Analyses of memory T cell populations demonstrated similar TCR diversity. Furthermore, experiments with individual mice demonstrated that primary effector and memory T cells have indistinguishable TCR repertoires. Remarkably, after reinfection with L. monocytogenes, LLO91-99-specific T cells have a narrower TCR repertoire than do primary effector or memory T cells. Thus, our studies show that the TCR repertoire of primary effector T lymphocytes is uniformly transmitted to memory T cells, whereas expansion of memory T cells is selective.


Subject(s)
Bacterial Toxins , Immunologic Memory/immunology , Listeria monocytogenes/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Bacterial Proteins/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Flow Cytometry , Heat-Shock Proteins/immunology , Hemolysin Proteins , Histocompatibility Antigens Class I/genetics , Humans , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Protein Conformation , Receptors, Antigen, T-Cell/genetics , Spleen/immunology
5.
J Exp Med ; 189(4): 701-10, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9989985

ABSTRACT

T lymphocyte recognition of infected cells is mediated by T cell receptors (TCRs) interacting with their ligands, self-major histocompatibility complex (MHC) molecules complexed with pathogen-derived peptides. Serial TCR interactions with potentially small numbers of MHC/ peptide complexes on infected cells transmit signals that result in T lymphocyte expansion and activation of effector functions. The impact of TCR affinity for MHC/peptide complexes on the rate or extent of in vivo T cell expansion is not known. Here we show that in vivo expansion of complex T cell populations after bacterial infection is accompanied by an increase in their overall affinity for antigen. T cell populations that have undergone additional rounds of in vivo expansion express a narrower range of TCRs, have increased sensitivity for antigen in cytotoxic T lymphocyte assays, and bind MHC/peptide complexes with greater affinity. The selective expansion of higher affinity T cells provides an in vivo mechanism for optimizing the early detection of infected cells.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Toxins , H-2 Antigens/immunology , Heat-Shock Proteins/immunology , Listeriosis/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Differentiation , Cell Division , Cells, Cultured , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Hemolysin Proteins , Immunization, Secondary , Immunologic Memory , Listeria monocytogenes/immunology , Mice , Mice, Inbred BALB C , Receptors, Antigen, T-Cell/immunology
6.
J Exp Med ; 190(2): 195-204, 1999 Jul 19.
Article in English | MEDLINE | ID: mdl-10432283

ABSTRACT

Major histocompatibility complex (MHC) class Ib molecules have been implicated in CD8(+) T cell-mediated defenses against intracellular bacterial infection, but the relative importance of MHC class Ib-restricted T cells in antimicrobial immunity is unknown. In this report, we use MHC tetramers to characterize T cell responses restricted by H2-M3, an MHC class Ib molecule that selectively presents N-formyl peptides. We find that sizeable H2-M3-restricted T cell responses, occurring earlier than MHC class Ia-restricted T cell responses, are mounted after primary infection with the intracellular bacterium Listeria monocytogenes. These H2-M3-restricted T cells are cytolytic and produce interferon gamma. However, after a second L. monocytogenes infection, H2-M3-restricted memory T cell responses are minor in comparison to the much larger MHC class Ia-restricted responses. This first direct characterization of an MHC class Ib-restricted T cell response indicates that CD8(+) T cells responding to L. monocytogenes infection can be divided into two groups: H2-M3-restricted responses, which provide rapid and quantitatively substantial effector function during primary infections but contribute relatively little to memory responses, and MHC class Ia-restricted responses, which expand later during primary infection but form memory T cells that respond rapidly and dramatically in response to subsequent infections by the same pathogen.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II , Listeriosis/immunology , Animals , Antigens, Bacterial/chemistry , Base Sequence , DNA Primers/genetics , Epitopes/chemistry , Female , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Immunologic Memory , In Vitro Techniques , Listeria monocytogenes/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Protein Conformation
7.
Cytogenet Genome Res ; 125(3): 186-200, 2009.
Article in English | MEDLINE | ID: mdl-19738379

ABSTRACT

The non-imprinted in Prader-Willi/Angelman syndrome (NIPA) proteins are highly conserved receptors or transporters. Translocation of NIPA genes were found in patients with Prader-Willi syndrome, and loss-of-function of the NIPA1 gene was identified in hereditary spastic paraplegia. The family of NIPA-like domain containing (NPAL) proteins is closely related to the NIPA proteins, but to date nothing is known about their function. Here, we could demonstrate that both human NPAL3 and mouse NPAL3 are ubiquitously expressed and encode highly conserved proteins. To further elucidate the function of the Npal3 gene, knockout (Npal3(-/-)) mice were generated. Intensive phenotypic analyses revealed that disruption of the Npal3 gene results in a pleiotropic phenotype. The function of the nervous system was impaired in both mutant males and females which could be demonstrated in behavioral tests. In addition, in NPAL3 mutants the number of NK cells was decreased and changes in IgM, IgG(2), and IgA were observed, indicating that the immune system is also affected. Interestingly, increased IgE levels as well as impaired lung functions were observed in mutant males but not in mutant females. It should be noted that the human Npal3 gene is located at 1p36.12-->p35.1, and atopic diseases were previously linked to this genomic region. Thus, the Npal3(-/-) mice could serve as a valuable model system for studying atopic diseases.


Subject(s)
Behavior, Animal , Immunoglobulin E/blood , Lung/physiology , Membrane Proteins/genetics , Amino Acid Sequence , Animals , Cation Transport Proteins , Cell Membrane/metabolism , Conserved Sequence , Evolution, Molecular , Female , Gene Expression , Humans , Immunoglobulin E/immunology , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Phenotype , Phylogeny , Sequence Alignment
8.
Rheumatology (Oxford) ; 47(3): 292-300, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18276736

ABSTRACT

OBJECTIVE: In a large-scale ENU (N-ethyl-N-nitrosourea) mouse mutagenesis programme, we previously have identified and characterized a novel mutation Ali18 that causes inflammatory arthritis like lesions in peripheral joints. In this study, we analysed the immune system of Ali18 mice to understand mechanisms underlying the spontaneous inflammation. METHODS: Humoral and cellular components of the immune system were phenotyped by ELISA and flow cytometry. The contribution of the immune system for phenotype expression was analysed in disease transfer experiments. The involvement of the adaptive immune system was investigated in Ali18;Rag1 double mutants and the influence of environmental factors was analysed in Ali18 mice reared under germ-free conditions. RESULTS: Bone marrow cells from Ali18 mice were able to transfer the disease phenotype to naïve wild-type recipients suggesting that cellular components of the reconstituted immune system were sufficient to induce arthritis. Ali18 mice revealed abnormal leucocyte populations including lymphocytes and granulocytes, as well as increased plasma IL-5 and IgE levels. Ali18;Rag1 double homozygous mutants, which lack mature lymphocytes, still developed arthritis, suggesting that the phenotype is independent of the adaptive immune system. In addition, the arthritis phenotype appeared to be independent from environmental conditions as demonstrated in mice reared under germ-free conditions. CONCLUSIONS: The Ali18 mutation induces inflammatory arthritis through bone marrow-derived cells. However, non-pro-inflammatory cytokine cascades and mature lymphocyte independent-mechanisms are crucial for initiation and progression of the phenotype. Ali18 mice may thus represent a model to study mechanisms involved in seronegative arthritis induced by cells of the innate immune system.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , B-Lymphocytes/immunology , T-Lymphocytes/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , B-Lymphocytes/metabolism , Bone Marrow Cells/immunology , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Probability , Random Allocation , Reference Values , Sensitivity and Specificity , T-Lymphocytes/metabolism
9.
Leukemia ; 31(10): 2161-2171, 2017 10.
Article in English | MEDLINE | ID: mdl-28090089

ABSTRACT

Cytomegalovirus (CMV) infection is a common, potentially life-threatening complication following allogeneic hematopoietic stem cell transplantation (allo-HSCT). We assessed prospectively the safety and efficacy of stem cell-donor- or third-party-donor-derived CMV-specific T cells for the treatment of persistent CMV infections after allo-HSCT in a phase I/IIa trial. Allo-HSCT patients with drug-refractory CMV infection and lacking virus-specific T cells were treated with a single dose of ex vivo major histocompatibility complex-Streptamer-isolated CMV epitope-specific donor T cells. Forty-four allo-HSCT patients receiving a T-cell-replete (D+ repl; n=28) or T-cell-depleted (D+ depl; n=16) graft from a CMV-seropositive donor were screened for CMV-specific T-cell immunity. Eight D+ depl recipients received adoptive T-cell therapy from their stem cell donor. CMV epitope-specific T cells were well supported and became detectable in all treated patients. Complete and partial virological response rates were 62.5% and 25%, respectively. Owing to longsome third-party donor (TPD) identification, only 8 of the 57 CMV patients transplanted from CMV-seronegative donors (D-) received antigen-specific T cells from partially human leukocyte antigen (HLA)-matched TPDs. In all but one, TPD-derived CMV-specific T cells remained undetectable. In summary, adoptive transfer correlated with functional virus-specific T-cell reconstitution in D+ depl patients. Suboptimal HLA match may counteract expansion of TPD-derived virus-specific T cells in D- patients.


Subject(s)
Cytomegalovirus Infections/therapy , Cytomegalovirus/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Immunotherapy, Adoptive/methods , T-Lymphocytes/transplantation , Viremia/therapy , Allografts , Antiviral Agents/therapeutic use , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/etiology , Cytomegalovirus Infections/transmission , Drug Resistance, Viral , Female , Graft Survival , Hematologic Neoplasms/therapy , Histocompatibility , Humans , Immunocompromised Host , Immunotherapy, Adoptive/adverse effects , Lymphocyte Depletion , Male , Myelodysplastic Syndromes/therapy , Prospective Studies , T-Cell Antigen Receptor Specificity , Tissue Donors , Viremia/drug therapy , Viremia/etiology
10.
Immunol Res ; 19(2-3): 211-23, 1999.
Article in English | MEDLINE | ID: mdl-10493175

ABSTRACT

Studies of the murine immune response to infection with the intracellular bacterial pathogen Listeria monocytogenes have provided a wealth of information about innate and acquired immune defenses in the setting of an infectious disease. Our studies have focused on the MHC class I restricted, CD8+ T cell responses of Balb/c mice to L. monocytogenes infection. Four peptides that derive from proteins that L. monocytogenes secretes into the cytosol of infected cells are presented to cytotoxic T lymphocyte (CTL) by the H2-Kd major histocompatibility complex (MHC) class I molecule. We have found that bacterially secreted proteins are rapidly degraded in the host cell cytosol by proteasomes that utilize, at least in part, the N-end rule to determine the rate of degradation. The MHC class I antigen processing pathway is remarkably efficient at generating peptides that bind to MHC class I molecules. The magnitude of in vivo T cell responses, however, is influenced to only a small degree by the amount of antigen or the efficiency of antigen presentation. Measurements of in vivo T cell expansion following L. monocytogenes infection indicate that differences in the sizes of peptide-specific T cell responses are more likely owing to differences in the repertoire of naive T cells than to differences in peptide presentation. This notion is supported by our additional finding that dominant T cell populations express a more diverse T cell receptor (TCR) repertoire than do subdominant T cell populations.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen Presentation , Epitopes , Listeria monocytogenes/pathogenicity , Mice , Receptors, Antigen, T-Cell
11.
Immunol Lett ; 65(1-2): 93-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10065633

ABSTRACT

Infection of mice with Listeria monocytogenes results in a robust T lymphocyte response that clears the pathogen and provides long term immunity from reinfection. The number of splenic CD4+ and CD8+ T cells and natural killer cells increases during primary and recall infection with L. monocytogenes, however the proportional increase is greatest for CD8+ T cells. The proportion of CD8 T cells expressing low levels of CD62L, a sign of activation, was increased among immune splenocytes, suggesting a substantial expansion of L. monocytogenes specific CTL. Analysis of CTL specific for the immunodominant LLO 91-99 epitope showed that essentially all were CD62Llo during the primary response, but that many upregulated expression of CD62L during the memory phase. Interestingly, the antigen specificity of nearly all additional CD62Llo CTL detected in spleens during recall L. monocytogenes infection can be accounted for with MHC class I tetramers complexed with four different epitopes. These studies demonstrate the complex T lymphocyte dynamics during infection with intracellular pathogens.


Subject(s)
Listeria monocytogenes/immunology , Listeriosis/immunology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , L-Selectin/metabolism , Listeriosis/microbiology , Mice , Mice, Inbred BALB C , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Cytotoxic/immunology
13.
Leukemia ; 28(12): 2355-66, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24736212

ABSTRACT

T cells have been proven to be therapeutically effective in patients with relapsed leukemias, although target antigens on leukemic cells as well as T-cell receptors (TCRs), potentially recognizing those antigens, are mostly unknown. We have applied an immunopeptidomic approach and isolated human leukocyte antigen (HLA) ligands from primary leukemia cells. We identified a number of ligands derived from different genes that are restrictedly expressed in the hematopoietic system. We exemplarily selected myeloperoxidase (MPO) as a potential target and isolated a high-avidity TCR with specificity for a HLA-B*07:02-(HLA-B7)-restricted epitope of MPO in the single HLA-mismatched setting. T cells transgenic for this TCR demonstrated high peptide and antigen specificity as well as leukemia reactivity in vitro and in vivo. In contrast, no significant on- and off-target toxicity could be observed. In conclusion, we here demonstrate, exemplarily for MPO, that leukemia-derived HLA ligands can be selected for specific effector tool development to redirect T cells to be used for graft manipulation or adoptive T-cell therapies in diverse transplant settings. This approach can be extended to other HLA ligands and HLA molecules in order to provide better treatment options for this life-threatening disease.


Subject(s)
HLA Antigens/immunology , Leukemia, Myeloid/genetics , Leukemia, Myeloid/immunology , Peptides/immunology , Peroxidase/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cell Survival/genetics , Cell Survival/immunology , Disease Models, Animal , Epitope Mapping , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , HLA Antigens/metabolism , HLA-B7 Antigen/immunology , HLA-B7 Antigen/metabolism , Heterografts , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/mortality , Ligands , Mice , Peptides/metabolism , Peroxidase/chemistry , Peroxidase/genetics , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity/immunology , Transduction, Genetic
14.
Blood Cancer J ; 1(3): e11, 2011 Mar.
Article in English | MEDLINE | ID: mdl-22829124

ABSTRACT

The FMS-like tyrosine kinase 3 (FLT3) is highly expressed in acute myeloid leukemia (AML). Internal tandem duplications (ITD) of the juxtamembrane domain lead to the constitutive activation of the FLT3 kinase inducing the activation of multiple genes, which may result in the expression of leukemia-associated antigens (LAAs). We analyzed the regulation of LAA in FLT3-wild-type (WT)- and FLT3-ITD(+) myeloid cells to identify potential targets for antigen-specific immunotherapy for AML patients. Antigens, such as PR-3, RHAMM, Survivin, WT-1 and PRAME, were upregulated by constitutively active FLT3-ITD as well as FLT3-WT activated by FLT3 ligand (FL). Cytotoxic T-cell (CTL) clones against PR-3, RHAMM, Survivin and an AML-directed CTL clone recognized AML cell lines and primary AML blasts expressing FLT3-ITD, as well as FLT3-WT(+) myeloid dendritic cells in the presence of FL. Downregulation of FLT3 led to the abolishment of CTL recognition. Comparing our findings concerning LAA upregulation by the FLT3 kinase with those already made for the Bcr-Abl kinase, we found analogies in the LAA expression pattern. Antigens upregulated by both FLT3 and Bcr-Abl may be promising targets for the development of immunotherapeutical approaches against myeloid leukemia of different origin.

15.
Curr Pharm Biotechnol ; 10(2): 236-43, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19199957

ABSTRACT

The German Mouse Clinic (GMC) is a large scale phenotyping center where mouse mutant lines are analyzed in a standardized and comprehensive way. The result is an almost complete picture of the phenotype of a mouse mutant line--a systemic view. At the GMC, expert scientists from various fields of mouse research work in close cooperation with clinicians side by side at one location. The phenotype screens comprise the following areas: allergy, behavior, clinical chemistry, cardiovascular analyses, dysmorphology, bone and cartilage, energy metabolism, eye and vision, host-pathogen interactions, immunology, lung function, molecular phenotyping, neurology, nociception, steroid metabolism, and pathology. The German Mouse Clinic is an open access platform that offers a collaboration-based phenotyping to the scientific community (www.mouseclinic.de). More than 80 mutant lines have been analyzed in a primary screen for 320 parameters, and for 95% of the mutant lines we have found new or additional phenotypes that were not associated with the mouse line before. Our data contributed to the association of mutant mouse lines to the corresponding human disease. In addition, the systemic phenotype analysis accounts for pleiotropic gene functions and refines previous phenotypic characterizations. This is an important basis for the analysis of underlying disease mechanisms. We are currently setting up a platform that will include environmental challenge tests to decipher genome-environmental interactions in the areas nutrition, exercise, air, stress and infection with different standardized experiments. This will help us to identify genetic predispositions as susceptibility factors for environmental influences.


Subject(s)
Biomedical Research/methods , Disease Models, Animal , Mice, Mutant Strains/genetics , Phenotype , Animal Husbandry , Animals , Biomedical Research/standards , Germany , Mice , Mice, Mutant Strains/growth & development , Quality Control
16.
Crit Care Med ; 10(4): 246-53, 1982 Apr.
Article in English | MEDLINE | ID: mdl-7067474

ABSTRACT

The feasibility of various metal chelates as components of acellular resuscitation fluids is examined, with particular emphasis on the bonding mode and kinetics of the dioxygen adduct formed. End-on (Pauling type) or bridged peroxo-dimer structures using iron(II) or cobalt(II) as the active metal ion are currently the most promising candidates for investigation. Criteria which must be met in order to confer desirable O2-binding properties upon the coordinated metal ions are described and ligand systems (porphyrins, Schiff-bases, lacunar macrobicycles) which help fulfill these criteria are critiqued. An important aspect of acellular O2-delivering systems is the mode of dispersion, which in turn is a function of the properties of the metal chelate; this question is broached, for general application, in terms of true solutions, emulsions and colloidally suspended solids.


Subject(s)
Blood Substitutes/therapeutic use , Chelating Agents/therapeutic use , Animals , Biological Transport , Chelating Agents/administration & dosage , Chelating Agents/metabolism , Cobalt/metabolism , Kinetics , Molecular Conformation , Oxygen/metabolism , Porphyrins/metabolism , Schiff Bases
17.
J Immunol ; 160(9): 4441-8, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9574549

ABSTRACT

Infection of BALB/c mice with Listeria monocytogenes primes CD8+ cytotoxic T cells specific for four different H2-Kd-restricted peptides. In vitro restimulation of L. monocytogenes immune splenocytes with each of these peptides resulted in larger T cell responses to p60 217-225 and mpl 84-92 than to LLO 91-99 and p60 449-457. Direct frequency analyses of immune splenocytes, however, revealed that LLO 91-99 and p60 217-225 elicit dominant T cell responses, while p60 449-457 and mpl 84-92 elicit minor, subdominant responses. Restimulation of immune splenocytes with a range of peptide concentrations revealed that T cells with dominant specificities respond optimally to low peptide concentrations, while T cells specific for subdominant epitopes expand maximally to high peptide concentrations. This disparity correlates with the stability of H2-Kd/epitope complexes: the two dominant epitopes form stable complexes, while the subdominant epitopes form less stable complexes with H2-Kd. Interestingly, T cells specific for LLO 91-99 and p60 217-225 express more complex TCR-Vbeta repertoires than p60 449-457- and mpl 84-92-specific T cells. Thus, in our system, dominant T cell responses have relatively diverse TCR repertoires and are specific for peptides that form stable complexes with MHC class I molecules. Determining the precise roles of epitope/MHC class I stability and TCR repertoire in the generation of dominant T cell responses will require further investigation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Animals , Antigen Presentation , Histocompatibility Antigens Class I/chemistry , Mice , Mice, Inbred BALB C , Peptides/chemistry , Peptides/immunology , Receptors, Antigen, T-Cell/immunology
18.
Rheumatol Int ; 17(4): 151-8, 1997.
Article in English | MEDLINE | ID: mdl-9440146

ABSTRACT

The objective of this study was to assess the validity of the lymphocyte proliferation assay in the diagnosis of Lyme arthritis (LA). We analyzed peripheral blood mononuclear cells of 11 patients with LA and 5 healthy controls for proliferative responses to live Borrelia burgdorferi by 3H-thymidine uptake. Before and after proliferation, the total number of cells was estimated and the relative amount of lymphocyte subsets was determined by FACS Lymphoproliferative responses to B. burgdorferi in patients with LA were predominantly due to CD4+ cells and in a small part due to CD8+ cells and were antigen-dependent in comparison to healthy donors. Considering the number of cells before proliferation, responses of TCR gamma/delta + cells and CD56+ cells were higher than responses to TCR alpha/beta+ and CD3+ cells in LA patients, but not in healthy donors. There were no demonstrable antigen-dependent CD19+ cell responses. Our results support the validity of lymphocyte proliferation assay in the diagnosis of LA.


Subject(s)
Borrelia burgdorferi Group/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lyme Disease/immunology , Lymphocyte Activation , T-Lymphocyte Subsets/immunology , Adolescent , Antigens, CD/analysis , Cells, Cultured , Child , Child, Preschool , Female , Humans , Leukocyte Count , Lyme Disease/diagnosis , Male , Middle Aged , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Reproducibility of Results , Thymidine/metabolism
19.
J Immunol ; 164(8): 4063-70, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10754299

ABSTRACT

Recent studies have demonstrated that viral and bacterial infections can induce dramatic in vivo expansion of Ag-specific T lymphocytes. Although presentation of Ag is critical for activation of naive T cells, it is less clear how dependent subsequent in vivo T cell proliferation and memory generation are upon Ag. We investigated T cell expansion and memory generation in mice infected alternately with strains of Listeria monocytogenes that contained or lacked an immunodominant, MHC class I-restricted T cell epitope. We found substantial differences in the responses of effector and memory T cells to inflammatory stimuli. Although effector T cells undergo in vivo expansion in response to bacterial infection in the absence of Ag, memory T cells show no evidence for such bystander activation. However, Ag-independent expansion of effector T cells does not result in increased memory T cell frequencies, indicating that Ag presentation is critical for effective memory T cell generation. Early reinfection of mice with L. monocytogenes before the maximal primary T cell response induces typical memory expansion, suggesting that the capacity for a memory T cell response exists within the primary effector population. Our findings demonstrate that T cell effector proliferation and memory generation are temporally overlapping processes with differing requirements for Ag.


Subject(s)
Antigens, Bacterial/physiology , Immunologic Memory/immunology , Listeria monocytogenes/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , Animals , Antigen Presentation , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Cell Division/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Inflammation/immunology , Inflammation/pathology , Kinetics , Listeria monocytogenes/genetics , Listeriosis/immunology , Listeriosis/pathology , Mice , Mice, Inbred BALB C , T-Lymphocyte Subsets/pathology
20.
J Immunol ; 166(2): 1132-40, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11145694

ABSTRACT

H2-M3-restricted presentation of N-formyl methionine (f-Met) peptides to CD8(+) T cells provides a mechanism for selective recognition of bacterial infection. In this report we demonstrate that Listeria monocytogenes infection induces distinct CD8(+) T cell populations specific for each of the known Listeria-derived formyl methionine peptides presented by M3. The sum H2-M3-restricted, Listeria-specific T cell response constitutes a major fraction of the total CD8(+) T cell response to primary infection. H2-M3-restricted T cell populations expand synchronously in vivo and achieve peak frequencies approximately 2 days earlier than MHC class Ia-restricted T cell populations. Although cross-recognition of different f-Met peptides by M3-restricted T cells was previously described, costaining of CD8(+) T cells ex vivo with H2-M3 tetramers complexed with different f-Met peptides shows that the majority of Listeria-specific, M3-restricted CD8(+) T cells are peptide specific. In contrast to the highly predictable size and immunodominance hierarchies of MHC class Ia-restricted T cell responses, the magnitudes of T cell responses specific for H2-M3-restricted peptides are remarkably variable between genetically identical mice. Our findings demonstrate that H2-M3-restricted T cell responses are distinct from classically restricted T cell responses to bacterial infection.


Subject(s)
Histocompatibility Antigens Class I/immunology , Immunodominant Epitopes/immunology , Immunodominant Epitopes/metabolism , Listeriosis/immunology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Animals , Cell Line , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Female , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Immunodominant Epitopes/chemistry , Kinetics , Listeria monocytogenes/immunology , Listeriosis/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , N-Formylmethionine , Peptide Fragments/chemistry , Protein Folding , Species Specificity , Spleen/chemistry , Spleen/cytology , Spleen/immunology , Staining and Labeling , T-Lymphocytes, Cytotoxic/chemistry , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL