Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
1.
J Intern Med ; 285(5): 510-523, 2019 05.
Article in English | MEDLINE | ID: mdl-30663801

ABSTRACT

Two related hyperinflammatory syndromes are distinguished following infection of humans with hantaviruses: haemorrhagic fever with renal syndrome (HFRS) seen in Eurasia and hantavirus pulmonary syndrome (HPS) seen in the Americas. Fatality rates are high, up to 10% for HFRS and around 35%-40% for HPS. Puumala virus (PUUV) is the most common HFRS-causing hantavirus in Europe. Here, we describe recent insights into the generation of innate and adaptive cell-mediated immune responses following clinical infection with PUUV. First described are studies demonstrating a marked redistribution of peripheral blood mononuclear phagocytes (MNP) to the airways, a process that may underlie local immune activation at the site of primary infection. We then describe observations of an excessive natural killer (NK) cell activation and the persistence of highly elevated numbers of NK cells in peripheral blood following PUUV infection. A similar vigorous CD8 Tcell response is also described, though Tcell responses decline with viraemia. Like MNPs, many NK cells and CD8 T cells also localize to the lung upon acute PUUV infection. Following this, findings demonstrating the ability of hantaviruses, including PUUV, to cause apoptosis resistance in infected target cells, are described. These observations, and associated inflammatory cytokine responses, may provide new insights into HFRS and HPS disease pathogenesis. Based on similarities between inflammatory responses in severe hantavirus infections and other hyperinflammatory disease syndromes, we speculate whether some therapeutic interventions that have been successful in the latter conditions may also be applicable in severe hantavirus infections.


Subject(s)
Adaptive Immunity , Hantavirus Pulmonary Syndrome/immunology , Hemorrhagic Fever with Renal Syndrome/immunology , Immunity, Innate , Puumala virus , Hantavirus Pulmonary Syndrome/therapy , Hemorrhagic Fever with Renal Syndrome/therapy , Humans , Severity of Illness Index
4.
Scand J Immunol ; 79(2): 90-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24313893

ABSTRACT

Anti-apoptotic proteins that block death receptor-mediated apoptosis favour tumour evasion of the immune system, leading to enhanced tumour progression. However, it is unclear whether blocking the mitochondrial pathway of apoptosis will protect tumours from immune cell attack. Here, we report that the anti-apoptotic protein Bcl-xL , known for its ability to block the mitochondrial pathway of apoptosis, exerted tumour-progressive activity in a murine lymphoma model. Bcl-xL overexpressing tumours exhibited a more aggressive development than control tumours. Surprisingly, Bcl-xL protection of tumours from NK cell-mediated attack did not involve protection from NK cell-mediated cytotoxicity. Instead, Bcl-xL -blocked apoptosis resulting from hypoxia and/or nutrient loss associated with the inhibition of angiogenesis caused by NK cell-secreted IFN-γ. These results support the notion that NK cells may inhibit tumour growth also by mechanisms other than direct cytotoxicity. Hence, the present results unravel a pathway by which tumours with a block in the mitochondrial pathway of apoptosis can evade the immune system.


Subject(s)
Interferon-gamma/physiology , Killer Cells, Natural/immunology , Lymphoma/immunology , Neovascularization, Pathologic/prevention & control , Tumor Escape , bcl-X Protein/physiology , Animals , Cell Line, Tumor , Cyclohexanes/pharmacology , Cytotoxicity, Immunologic , Humans , Mice , Mice, Inbred C57BL , O-(Chloroacetylcarbamoyl)fumagillol , Sesquiterpenes/pharmacology
5.
Scand J Immunol ; 76(6): 559-66, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22928727

ABSTRACT

The differentiation and maintenance of Th17 cells require a unique cytokine milieu and activation of lineage-specific transcription factors. This process appears to be antagonized by the transcription factor T-bet, which controls the differentiation of Th1 cells. Considering that T-bet-deficient (T-bet(-/-) ) mice are largely devoid of natural killer (NK) cells due to a defect in the terminal maturation of these cells, and because NK cells can influence the differentiation of T helper cells, we investigated whether the absence of NK cells in T-bet-deficient mice contributes to the augmentation of autoreactive Th17 cell responses. We show that the loss of T-bet renders the transcription factors Rorc and STAT3 highly responsive to activation by stimuli provided by NK cells. Furthermore, reconstitution of T-bet(-/-) mice with wild-type NK cells inhibited the development of autoreactive Th17 cells through NK cell-derived production of IFN-γ. These results identify NK cells as critical regulators in the development of autoreactive Th17 cells and Th17-mediated pathology.


Subject(s)
Killer Cells, Natural/immunology , T-Box Domain Proteins/metabolism , Th17 Cells/immunology , Animals , Autoantigens/immunology , Cell Communication/genetics , Cell Communication/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Cytokines/immunology , Cytotoxicity, Immunologic , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Immune Tolerance , Killer Cells, Natural/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , T-Box Domain Proteins/genetics , Th1 Cells/immunology
6.
Leukemia ; 24(9): 1607-16, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20613786

ABSTRACT

Myelodysplastic syndromes (MDS) comprise a heterogeneous group of clonal stem-cell disorders characterized by ineffective hematopoiesis and risk of progression to acute myeloid leukemia. Increased apoptosis and suppressed functions of peripheral blood natural killer (NK) cells have been described in MDS patients, but only limited information is available on the phenotypic and functional integrity of NK cells in the bone marrow. In a cohort of 41 patients with distinct clinical subtypes of MDS, we here show that NK cells in the bone marrow show decreased surface expression of the activating receptors DNAM-1 and NKG2D. Notably, decreased receptor expression correlated with elevated bone marrow blast counts and was associated with impaired NK-cell responsiveness to stimulation with the K562 cell line, or co-activation by NKG2D or DNAM-1 in combination with the 2B4 receptor. Furthermore, antibody-masking experiments revealed a central role for DNAM-1 in NK cell-mediated killing of freshly isolated MDS blasts. Thus, given the emerging evidence for NK cell-mediated immune surveillance of neoplastic cells, we speculate that reduced expression of DNAM-1 on bone marrow NK cells may facilitate disease progression in patients with MDS.


Subject(s)
Antigens, CD34/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , Apoptosis , Bone Marrow Cells/metabolism , Killer Cells, Natural/metabolism , Myelodysplastic Syndromes/metabolism , Adult , Aged , Aged, 80 and over , Bone Marrow Cells/immunology , Disease Progression , Female , Flow Cytometry , Humans , Killer Cells, Natural/immunology , Male , Middle Aged , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/pathology
8.
J Med Genet ; 45(3): 134-41, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17993578

ABSTRACT

OBJECTIVE: Familial haemophagocytic lymphohistiocytosis (FHL) is a fatal disorder of immune dysregulation with defective cytotoxic lymphocyte function. Disease-causing mutations have been identified in the genes encoding perforin (PRF1), syntaxin-11 (STX11), and Munc13-4 (UNC13D). We screened for UNC13D mutations and studied clinical and functional implications of such mutations in a well defined patient cohort. METHODS: Sequencing of UNC13D was performed in 38 FHL patients from 34 FHL families in which PRF1 and STX11 mutations had been excluded. RESULTS: We identified six different mutations affecting altogether 9/38 individuals (24%) in 6/34 (18%) unrelated PRF1/STX11-negative families. Four novel mutations were revealed; two homozygous nonsense mutations (R83X and W382X), one splice mutation (exon 28), and one missense mutation (R928P). In addition, two known mutations were identified (R214X and a deletion resulting in a frame-shift starting at codon 782). There was considerable variation in the age at diagnosis, ranging from time of birth to 14 years (median 69 days). Three of nine patients (33%) developed central nervous system (CNS) symptoms. Natural killer (NK) cell activity was impaired in all four patients studied. Defective cytotoxic lymphocyte degranulation was evident in the two patients investigated, more pronounced in the patient with onset during infancy than in the patient with adolescent onset. CONCLUSIONS: Biallelic UNC13D mutations were found in 18% of the PRF1/STX11-negative FHL families. Impairment of NK cell degranulation was less pronounced in a patient with adolescent onset. FHL should be considered not only in infants but also in adolescents, and possibly young adults, presenting with fever, splenomegaly, cytopenia, hyperferritinaemia, and/or CNS symptoms.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/genetics , Membrane Proteins/genetics , Mutation , Adolescent , Age of Onset , Cell Degranulation , Child , Child, Preschool , Codon, Nonsense/genetics , Female , Frameshift Mutation , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Killer Cells, Natural/immunology , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/immunology , Male , Membrane Proteins/immunology , Mutation, Missense , Perforin , Pore Forming Cytotoxic Proteins/genetics , Qa-SNARE Proteins/genetics , Sequence Deletion
9.
Scand J Immunol ; 65(4): 353-61, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17386026

ABSTRACT

Peripheral neurones have the potential to transmit infectious agents to the central nervous system (CNS). This raises the possibility of existing host defence mechanisms that may prevent such spread. Natural killer (NK) cells can target infected cells, and by this ability serve to limit spread of infection prior to the development of adaptive immune responses. To address directly if NK cells can target infected peripheral neurones, we examined the expression of NK cell-activating ligands and susceptibility to NK cell-mediated cytolytic effects in ex vivo cultures of mouse peripheral dorsal root ganglia (DRG) neurones prior to and after infection with a neurotropic strain of influenza A virus, WSN/33. In infected DRG cultures, retinoic acid early inducible gene-1 (RAE-1) transcripts were induced and exposure to interleukin (IL)-2-activated NK cells resulted in a total destruction of neurites. Studies on cultures from interferon (IFN)-alpha/betaR-deficient mice suggest that the infection engages an IFN-alpha/beta-dependent signalling pathway to induce RAE-1 transcripts. In contrast, induction of RAE-1 transcripts or NK cell-mediated neurite destructions was not observed in central hippocampal neurones. This reveals distinct properties between peripheral DRG and central hippocampal neurones with respect to the ability to signal for immune destruction following infection.


Subject(s)
Ganglia, Spinal/virology , Hippocampus/virology , Killer Cells, Natural/immunology , Neurons/virology , Orthomyxoviridae Infections/immunology , Animals , Cells, Cultured , Cytotoxicity, Immunologic , Ganglia, Spinal/immunology , Hippocampus/immunology , Immunohistochemistry , Influenza A virus/immunology , Interferon-alpha/immunology , Interferon-alpha/metabolism , Interferon-beta/immunology , Interferon-beta/metabolism , Male , Mice , Orthomyxoviridae Infections/transmission , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/metabolism
10.
Leukemia ; 20(5): 833-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16511516

ABSTRACT

The possibility of using natural killer (NK) cells in treatment of human hematological malignancies has increased in recent years. One factor contributing to this is the introduction of new methods for ex vivo generation of enriched populations of clinical grade NK cells. The objective of the present study was to evaluate the safety and efficacy of human ex vivo expanded clinical grade NK cells against K562 leukemia cells in severe combined immunodeficiency disease (SCID)-beige mice. Irradiated SCID-beige mice were injected intravenously (i.v.) with K562 leukemia cells. Following leukemia cell injection, mice were injected with ex vivo expanded human NK cells. NK cells were followed in vivo and mice monitored for survival from leukemia. Administration of these ex vivo expanded clinical grade NK cells was safe and prevented leukemia development. In conclusion, these results imply possibilities for the use of this NK cell preparation in treatment trials of human hematological malignancies and possibly other forms of cancer.


Subject(s)
Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Leukemia, Experimental/immunology , Leukemia, Experimental/therapy , Lymphocyte Transfusion/methods , Animals , Cell Transplantation , Cytotoxicity, Immunologic , Disease Models, Animal , Flow Cytometry , Graft vs Host Disease/prevention & control , Humans , In Vitro Techniques , Injections, Intraperitoneal , K562 Cells , Killer Cells, Natural/cytology , Leukemia, Experimental/genetics , Mice , Mice, SCID , Neoplasm Transplantation , Phenotype , Transplantation, Heterologous , Tumor Cells, Cultured
11.
Scand J Immunol ; 62(5): 437-44, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16305640

ABSTRACT

In subjects not developing allergy, inhalation of nonpathogenic protein antigens causes no harm and is associated with tolerance induction. Repeated exposure to aerosolized ovalbumin (OVA) likewise does not evoke airway inflammation and induces inhalation tolerance in experimental animals. The present study explored the role of the inhibitory T-cell receptor CTLA-4, in preventing inflammation and in establishing inhalation tolerance in response to a protein antigen. Naive BALB/c mice were injected intraperitoneally with anti-CTLA-4 monoclonal antibody or control immunoglobulin G (IgG) and exposed daily to aerosolized saline or OVA over 10 or 20 consecutive days. OVA-specific IgE levels and the inflammatory response in airway tissues were assessed 2 days after last exposure. The OVA-specific IgE response was also evaluated in mice subjected to a subsequent immunogenic OVA challenge 18 days after last aerosol exposure. Additional mice were made tolerant by 10 days of OVA aerosol exposure and were then subjected to an immunogenic OVA challenge combined with CTLA-4 blockade or control IgG treatment. Repeated inhalation of aerosolized OVA alone did not cause a pulmonary inflammatory response. In contrast, 10 days of OVA exposure combined with blockade of CTLA-4 led to development of eosinophilic lung infiltrates, BAL fluid eosinophilia, goblet cell hyperplasia and increased OVA-specific IgE. By 20 days of OVA exposure and blockade of CTLA-4, the inflammatory response remained. Mice exposed to aerosolized OVA for 10 days exhibited greatly reduced OVA-specific IgE responses to subsequent immunogenic OVA challenge. Blockade of CTLA-4 during the period of OVA aerosol exposure did not prevent this suppression of the OVA-specific IgE response. Neither did blockade of CTLA-4 during immunogenic OVA challenge alter the allergen-specific IgE response. Our results indicate that in vivo blockade of CTLA-4 modulates the initial immune response to a protein antigen allowing the development of allergen-induced airway inflammation in naive mice. However, this initial exaggerated immune response is followed by the induction of inhalation tolerance, demonstrating that CTLA-4 signalling is not decisive in this process. Our findings also show that once inhalation tolerance is established it may not be disrupted by blockade of CTLA-4.


Subject(s)
Antigens, Differentiation/immunology , Immune Tolerance/immunology , Immunity, Mucosal/immunology , Pneumonia/immunology , Aerosols , Allergens/administration & dosage , Allergens/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antigens, CD , Bronchoalveolar Lavage Fluid/cytology , CTLA-4 Antigen , Eosinophils/cytology , Female , Immunity, Mucosal/drug effects , Immunoglobulin E/blood , Immunoglobulin E/immunology , Injections, Intraperitoneal , Lung/cytology , Lung/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Ovalbumin/immunology , Pneumonia/chemically induced , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology
12.
Scand J Immunol ; 60(1-2): 14-22, 2004.
Article in English | MEDLINE | ID: mdl-15238069

ABSTRACT

Natural killer (NK) cells, the third major lymphocyte population, are important effector cells against certain infections and tumours. They have also been implicated as a link between innate and adaptive immune responses. In recent years, much attention has been paid to the NK cell inhibitory receptors and their interaction with major histocompatibility complex class I molecules on target cells. This review summarizes recent findings on regulation of NK cell activity with an emphasis on NK cell stimulatory receptors. A particular emphasis is devoted to the receptor NKG2D that is expressed on all NK cells.


Subject(s)
Killer Cells, Natural/immunology , Receptors, Immunologic/immunology , Animals , Antigens, CD/immunology , Antigens, Ly/immunology , Histocompatibility Antigens Class I/immunology , Humans , Lectins, C-Type/immunology , Ligands , Lymphocyte Activation/immunology , Mice , NK Cell Lectin-Like Receptor Subfamily D , NK Cell Lectin-Like Receptor Subfamily K , Receptors, KIR , Receptors, NK Cell Lectin-Like , Receptors, Natural Killer Cell
13.
Leukemia ; 17(10): 1973-80, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14513047

ABSTRACT

B-cell chronic lymphocytic leukemia (B-CLL) is the most common leukemia in the Western world. It is currently an incurable disease, making new treatment options such as immunotherapy desirable. Monoclonal antibodies (Mabs) to surface antigens of the tumor cell is one option. Administration of cytotoxic cells such as natural killer (NK) and natural killer-like T (NKT) cells expanded in vitro might be a useful treatment modality alone or in combination with MAbs. A limiting step in the development of successful cellular immunotherapy has been the availability of appropriate cytotoxic cells. Here, we report the feasibility of expanding populations of the human killer cells, CD3-CD56+ NK and CD3+CD56+ NKT cells, from peripheral blood mononuclear cells (PBMCs) of B-CLL patients. The influence of tumor B cells on the in vitro expansion of killer cells was assessed by depleting B cells from PBMCs by microbead separation before culture. The 21-day cultures from both B-cell- and non-B-cell-depleted PBMC showed a marked expansion of NK cells, and also of T cells, among which almost half had the NKT phenotype. Depletion of B cells before culture did not change the expansion rates of NK and NKT cells significantly. In patients with progressive B-CLL, NK cell expansion capacity was improved after fludarabine treatment when compared to samples obtained before treatment. Repeated samples of PBMCs from individual untreated patients with both indolent and progressive disease cultured under identical conditions gave similar NK cell expansion rates. Expanded killer cell populations had cytotoxic function against the NK-sensitive target K562 cell line and expressed high levels of Granzyme B. From our studies, we conclude that NK cells as well as NKT cells from the peripheral blood of B-CLL patients can be expanded, and that these cells have cytotoxic capacity.


Subject(s)
Killer Cells, Natural/immunology , Leukemia, B-Cell/immunology , T-Lymphocytes/immunology , Aged , Antigens, CD/blood , Cytotoxicity, Immunologic , Female , Flow Cytometry , Humans , Immunotherapy/methods , Leukemia, B-Cell/therapy , Lymphocyte Count , Lymphocyte Depletion , Male , Middle Aged , T-Lymphocyte Subsets/immunology , T-Lymphocytes/classification
14.
Scand J Immunol ; 55(5): 432-41, 2002 May.
Article in English | MEDLINE | ID: mdl-11975754

ABSTRACT

Autoimmune diseases are chronic conditions resulting from a loss of immunological tolerance to self-antigens. Recent observations have supported an ever-broader role for innate immune responses in directing and regulating adaptive immunity, including responses to self. This review summarizes recent findings supporting important functions of natural killer (NK) cells in regulating autoimmunity. A close survey of the current literature reveals multiple steps where NK cells can regulate inflammation and intervene in loss of self-tolerance. Importantly, the findings also caution against inferring a similar role for NK cells in all autoimmune phenomena or during separate stages of the same disease. Indeed, NK cells may have different influences during the priming and the effector phases of disease. Hence, an increased understanding of the involvement of NK cells in inflammation and infection should provide new insights into the pathogenesis of autoimmune disease.


Subject(s)
Autoimmune Diseases/immunology , Killer Cells, Natural/immunology , Animals , Autoantigens/immunology , Humans , Immune Tolerance/immunology , Mice
15.
Eur J Immunol ; 31(11): 3248-54, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11745341

ABSTRACT

CD8(+) T cells are known to down-regulate the TCR complex upon ligation with its cognate MHC class I-peptide complex. In the present report, we demonstrate that stimulation of CD8(+) T cells with cytokines also leads to down-regulation of the TCR complex and TCR-associated surface molecules. A significant reduction of TCRalpha beta, CD3, CD8alpha and CD8beta surface expression was observed when CD8(+) T cells were cultured in IL-2 and to a lesser extent in IL-4 or IL-15. The down-regulation was apparent after 2 days of culture and was observed at IL-2 concentrations as low as 10 U/ml. Using TCR transgenic mice, we found that the down-regulation was associated with a decreased affinity of CD8(+) T cells to MHC class I-peptide complexes, as determined by MHC class I tetramer staining. Furthermore, the antigen-specific proliferation of IL-2-pre-activated CD8(+) T cells was significantly reduced compared to naive CD8(+) T cells or to CD8(+) T cells previously stimulated with peptide-pulsed dendritic cells. Moreover, only CD8alpha(high) but not CD8alpha(low) cells sorted from IL-2-activated CD8(+) T cells proliferated in response to specific antigen, although both subsets proliferated equally well to IL-2. Taken together, these data suggest that the down-regulation of TCR components and a subsequent decrease in affinity towards MHC class I-peptide complexes may be a mechanism by which TCR-dependent proliferation of non-specifically activated CD8(+) T cells is avoided.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Interleukin-2/pharmacology , Receptors, Antigen, T-Cell/analysis , Animals , CD8 Antigens/analysis , CD8-Positive T-Lymphocytes/chemistry , Down-Regulation , Histocompatibility Antigens Class I/metabolism , Interleukin-15/pharmacology , Interleukin-4/pharmacology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Receptor-CD3 Complex, Antigen, T-Cell/analysis , Receptors, Antigen, T-Cell, alpha-beta/analysis
16.
J Immunol ; 167(12): 6706-10, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739483

ABSTRACT

The biological function of 2B4, a CD48-binding molecule expressed on T cells with an activation/memory phenotype, is not clear. In this report, we demonstrate that proliferation of CD8(+) T cells is regulated by 2B4. Proliferative responses of CD8(+) T cells were significantly reduced by anti-2B4 Ab. The effects were not potentiated by anti-CD48 Ab, suggesting that the observed responses were driven by 2B4/CD48 interactions. Surprisingly, the 2B4/CD48-dependent proliferative responses were also observed in the absence of APCs. This suggests that 2B4/CD48 interactions can occur directly between T cells. Furthermore, when activated 2B4(+)CD8(+) T cells were mixed with 2B4(-)CD8(+) TCR-transgenic T cells and specific peptide-loaded APC, the proliferation of the latter T cells was inhibited by anti-2B4 Ab. Taken together, this suggests that 2B4 on activated/memory T cells serves as a ligand for CD48, and by its ability to interact with CD48 provides costimulatory-like function for neighboring T cells.


Subject(s)
Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Membrane Glycoproteins/metabolism , Receptors, Immunologic , Animals , Antibodies/pharmacology , Antigen-Presenting Cells/immunology , Antigens/immunology , Antigens, CD/immunology , CD48 Antigen , Cells, Cultured , Cytokines/pharmacology , Flow Cytometry , Genes, T-Cell Receptor , Immunologic Memory , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/biosynthesis , Signaling Lymphocytic Activation Molecule Family
17.
J Leukoc Biol ; 70(6): 903-10, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11739553

ABSTRACT

The administration of cAMP-elevating agents affects a number of autoimmune and inflammatory conditions. Because dendritic cells (DCs) play a pivotal role in autoimmunity and inflammation, the isolated effects of cAMP-elevating agents on the function of DCs was examined. In a dose-dependent manner, 8-Bromo cAMP, prostaglandin E(2), and 3-isobutyl-1-methylxanthine inhibited tumor necrosis factor alpha release and suppressed antigen presentation by DCs. The same effect was observed with rolipram, a specific inhibitor of phosphodiesterase type 4, but not with inhibitors of other phosphodiesterases. The decreased antigen presentation by DCs was associated with an enhanced production of interleukin (IL)-10 and with lower major histocompatibility complex type II (MHC II) expression. Furthermore, the inhibition of antigen presentation and MHC II expression was significantly reversed by treatment of DCs with neutralizing antibody against IL-10, suggesting the involvement of an IL-10-dependent mechanism. Taken together, these results might explain why certain cAMP-elevating agents such as rolipram are effective in blocking autoimmunity and inflammation.


Subject(s)
Antigen Presentation/drug effects , Cyclic AMP/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cells, Cultured , Interleukin-10/immunology , Mice , Mice, Inbred BALB C , Phosphodiesterase Inhibitors/pharmacology , Rolipram/pharmacology , Tumor Necrosis Factor-alpha/immunology
18.
Hum Immunol ; 62(10): 1092-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11600215

ABSTRACT

Adoptive transfer of immunocompetent cells may induce anti-tumor effects in vivo. However, a significant obstacle to the development of successful cellular immunotherapy has been the availability of appropriate cytotoxic cells. Among the immunologic effector cells that are considered mediators of anti-tumor effects, those with the highest per-cell cytotoxic capacity express a natural killer (NK) cell phenotype, i.e., CD56(+)CD3(-). However, such cells are normally present only in low numbers in peripheral blood mononuclear cells (PBMCs), lymphokine activated killer (LAK), and cytokine induced killer (CIK) cell preparations. To optimize the expansion of human NK cells, PBMCs were cultured in different serum free medium supplemented with monoclonal anti-CD3 antibodies and interleukin (IL)-2 at varying concentrations. By using Cellgro stem cell growth medium supplemented with 5% human serum and IL-2 (500 U/ml) cells expanded 193-fold (median, range 21-277) after 21 days, and contained 55% (median, range 7-92) CD3(-)CD56(+) cells. The remaining cells were CD3(+) T cells, 22% (median, range 2-68) of which co-expressed CD56. The expanded cell population lysed 26 to 45% of K562 targets in a 1:1 effector to target ratio, signifying substantial cytotoxic efficacy. The described method is a simple and efficient way of expanding and enriching human NK cells. We have termed these high-yield CD3(-)CD56(+) cells cytokine-induced natural killer (CINK) cells.


Subject(s)
CD3 Complex/biosynthesis , CD56 Antigen/biosynthesis , Cell Culture Techniques/methods , Cytotoxicity, Immunologic , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocyte Activation , Cell Division/immunology , Cell Separation , Cells, Cultured , Centrifugation, Density Gradient , Culture Media, Serum-Free , Cytotoxicity Tests, Immunologic , Humans , Immunophenotyping , Immunotherapy, Adoptive , Interleukin-2/pharmacology , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism
19.
Clin Exp Allergy ; 31(10): 1583-93, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11678859

ABSTRACT

BACKGROUND: The yeast Malassezia furfur (M. furfur), present in the normal microflora of human skin, can act as an allergen that incites specific IgE reactivity and T cell proliferation in atopic dermatitis (AD) patients. The role of antigen presenting dendritic cells (DCs) in the onset and maintenance of AD is not well established. OBJECTIVE: The objective of the present study was to assess whether the interaction of M. furfur with human DCs will result in DC maturation, cytokine production and lymphocyte proliferation. METHODS: Monocyte-derived dendritic cells (MDDCs) were generated from human peripheral blood. Immature MDDCs were cultured with or without M. furfur or plastic beads, and with or without CD40L stimulation. Interaction of yeast cells by MDDCs was studied by time-lapse photography and cytokines were detected in culture supernatants with ELISA. The ability of MDDCs pre-incubated with M. furfur to induce proliferation in autologous lymphocytes was measured by [(3)H]-thymidine incorporation. RESULTS: Time-lapse photography showed that the majority of immature MDDCs internalized whole M. furfur yeast cells within 1 h. The presence of M. furfur induced maturation (CD83 expression) of MDDCs, and up-regulation of the costimulatory molecules CD80 and CD86. Production of TNF-alpha, IL-1 beta and IL-18 by MDDCs increased significantly (P < 0.05 for TNF-alpha and IL-1 beta, and P < 0.01 for IL-18) after the addition of M. furfur, while IL-10 and IL-12p70 levels remained unaltered. The CD40L-stimulated IL12p70 production by MDDCs was decreased in the presence of M. furfur (P < 0.05). Finally, immature MDDCs pre-incubated with M. furfur induced a proliferative response in autologous CD14-depleted peripheral blood mononuclear cells, in a dose-dependent manner. CONCLUSION: The data indicate that immature MDDCs can internalize the opportunistic yeast M. furfur. This process was associated with MDDC maturation, production of pro-inflammatory and immunoregulatory cytokines, which might favour induction of a Th2-type immune response, and a capacity to stimulate lymphocyte proliferation. This chain of events most likely contributes to the inflammatory reaction in AD.


Subject(s)
Allergens/adverse effects , Dendritic Cells/cytology , Malassezia , Yeasts , Antigens, CD/physiology , CD40 Ligand/biosynthesis , Cell Division/immunology , Cells, Cultured , Coculture Techniques , Cytokines/biosynthesis , Humans , Lymphocyte Activation/immunology , Lymphocytes/immunology , Pilot Projects , Reference Values , Time Factors
20.
J Immunol ; 167(4): 2068-73, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11489989

ABSTRACT

NK cells provide a line of defense against tumors and virus-infected cells that have lost the expression of one or more MHC class I isoforms. Here, we investigate whether inhibitors of apoptosis can block the rejection of tumors mediated by NK cells, by introducing the long form of Fas-associated death domain-like IL-1beta-converting enzyme-associated inhibitory protein (FLIP(L)) and poxvirus cytokine response modifier A (CrmA) into the MHC class I-deficient T lymphoma cell line RMA-S. RMA-S cells do not normally express Fas in vitro, and it was previously postulated that the rejection of these tumors by NK cells is strictly perforin dependent. We show that perforin-deficient NK cells directly mediate Fas up-regulation on RMA-S cells and thereafter kill the cells in a Fas-dependent manner, and that RMA-S FLIP(L) and RMA-S CrmA are protected from such killing. When injected in immunocompetent recipients, RMA-S cells up-regulate Fas, rendering in vivo-passed mock-transduced cells sensitive to Fas-mediated apoptosis. Moreover, RMA-S FLIP(L) and RMA-S CrmA cells establish aggressive tumors, in contrast to RMA-S mock cells that are rejected. These results demonstrate that FLIP(L) and CrmA function as tumor progression factors by protecting MHC class I-deficient tumors from rejection mediated by NK cells. Moreover, our data indicate that death receptor-mediated apoptosis has a more prominent role in the clearance of NK-sensitive tumors than previously suggested.


Subject(s)
Apoptosis/immunology , Carrier Proteins/physiology , Graft Rejection/immunology , Intracellular Signaling Peptides and Proteins , Killer Cells, Natural/immunology , Serpins/physiology , Animals , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/genetics , Cysteine Proteinase Inhibitors/pharmacology , Cytotoxicity, Immunologic/genetics , Disease Progression , Genetic Vectors/immunology , Graft Rejection/genetics , Graft Rejection/pathology , Humans , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/pathology , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Perforin , Pore Forming Cytotoxic Proteins , Poxviridae/genetics , Serpins/genetics , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/transplantation , Up-Regulation/immunology , Viral Proteins/genetics , fas Receptor/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...