Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Photochem Photobiol ; 83(5): 1095-100, 2007.
Article in English | MEDLINE | ID: mdl-17880504

ABSTRACT

Exposing experimental animals or human volunteers to UVA II (320-340 nm) radiation after immunization suppresses immunologic memory and the elicitation of delayed-in-time hypersensitivity reactions. Previous studies indicated that the mechanisms underlying UVA-induced immune suppression are similar to those described for UVB-induced immune suppression, i.e. transferred by T regulatory cells, overcome by repairing DNA damage, neutralizing interleukin (IL)-10 activity, or injecting recombinant IL-12. Here we continued our examination of the mechanisms involved in UVA II-induced suppression. Antibodies to cis-urocanic acid blocked UVA-induced immune suppression. Treating UVA-irradiated mice with histamine receptor antagonists, calcitonin gene-related peptide (CGRP) receptor antagonists or platelet activating factor receptor antagonists blocked immune suppression in UVA-irradiated mice. In light of the fact that cis-urocanic acid and CGRP target mast cells, which can then release platelet activating factor and histamine, we measured UVA-induced immune suppression in mast cell-deficient mice. No immune suppression was noted in UVA-irradiated mast cell-deficient mice. These findings indicate that exposure to UVA II activates many of the same immune regulatory factors activated by UVB to induce immune suppression. Moreover, they indicate that mast cells play a critical role in UVA-induced suppression of secondary immune reactions.


Subject(s)
Mast Cells/radiation effects , Ultraviolet Rays , Animals , Calcitonin Gene-Related Peptide/physiology , Histamine H1 Antagonists/pharmacology , Mast Cells/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors
2.
Proc Natl Acad Sci U S A ; 103(46): 17420-5, 2006 Nov 14.
Article in English | MEDLINE | ID: mdl-17085585

ABSTRACT

Exposure to UV radiation induces skin cancer and suppresses the immune response. To induce immune suppression, the electromagnetic energy of UV radiation must be absorbed by an epidermal photoreceptor and converted into a biologically recognizable signal. Two photoreceptors have been recognized: DNA and trans-urocanic acid (UCA). Trans-UCA is normally found in the outermost layer of skin and isomerizes to the cis isomer upon exposure to UV radiation. Although UCA was identified as a UV photoreceptor years ago, and many have documented its ability to induce immune suppression, its exact mode of action remains elusive. Particularly vexing has been the identity of the molecular pathway by which cis-UCA mediates immune suppression. Here we provide evidence that cis-UCA binds to the serotonin [5-hydroxytryptamine (5-HT)] receptor with relatively high affinity (Kd = 4.6 nM). Anti-cis-UCA antibody precipitates radiolabeled 5-HT, and the binding is inhibited by excess 5-HT and/or excess cis-UCA. Similarly, anti-5-HT antibody precipitates radiolabeled cis-UCA, and the binding is inhibited by excess 5-HT or excess cis-UCA. Calcium mobilization was activated when a mouse fibroblast line, stably transfected with the human 5-HT2A receptor, was treated with cis-UCA. Cis-UCA-induced calcium mobilization was blocked with a selective 5-HT2A receptor antagonist. UV- and cis-UCA-induced immune suppression was blocked by antiserotonin antibodies or by treating the mice with 5-HT2A receptor antagonists. Our findings identify cis-UCA as a serotonin receptor ligand and indicate that the immunosuppressive effects of cis-UCA and UV radiation are mediated by activation of the 5-HT2A receptor.


Subject(s)
Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Photosensitizing Agents/pharmacology , Receptor, Serotonin, 5-HT2A/metabolism , Sunlight , Urocanic Acid/pharmacology , Animals , Binding Sites , Calcium/metabolism , Cell Line , Humans , Immunosuppressive Agents/chemistry , Mice , Molecular Structure , Photosensitizing Agents/chemistry , Serotonin Antagonists/pharmacology , Spodoptera , Urocanic Acid/chemistry
3.
J Immunol ; 177(7): 4810-7, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16982922

ABSTRACT

UV exposure suppresses the immune response to a variety of microbial, fungal, and viral Ags. In addition, UV radiation is a complete carcinogen and the immune suppression induced by UV radiation is a major risk factor for skin cancer induction. In this study, we examined the mechanisms underlying the induction of immune suppression and tolerance induction by UV radiation. Transferring lymph nodes cells from UV-irradiated, FITC-sensitized mice into normal recipients transferred immune tolerance. Contrary to expectations, the cell responsible was an FITC(+), IL-10-secreting, CD19(+), B220(+) B cell. Because the lipid mediator of inflammation, platelet-activating factor (PAF) is released by UV-irradiated keratinocytes and is essential for the induction of immune suppression, we determined its role in tolerance induction. When UV-irradiated mice were injected with PCA 4248, a selective PAF receptor (PAFR) antagonist, transfer of tolerance was suppressed. However, immune suppression was not transferred when FITC(+) cells from the draining lymph nodes of UV-irradiated, PAFR-deficient donor mice were injected into the recipients. Because PCA 4248 also blocks serotonin receptor binding, we measured the effect that blocking both serotonin and PAFR binding has on the transfer of immune suppression. Only when both PAF and serotonin binding were blocked could we inhibit tolerance induction. These data identify a novel function for PAF and serotonin in modulating immune function, the activation of immunoregulatory B cells.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/radiation effects , Dermatitis, Contact/immunology , Immune Tolerance , Platelet Activating Factor/metabolism , Serotonin/metabolism , Adoptive Transfer , Animals , Antigens, CD19/metabolism , Dihydropyridines/pharmacology , Flow Cytometry , Fluorescein-5-isothiocyanate , Inflammation/immunology , Interleukin-10/metabolism , Leukocyte Common Antigens/metabolism , Lymphocyte Activation/immunology , Lymphocyte Activation/radiation effects , Mice , Mice, Mutant Strains , Platelet Activating Factor/antagonists & inhibitors , Serotonin Antagonists/pharmacology , Ultraviolet Rays
4.
Am J Pathol ; 169(3): 795-805, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16936256

ABSTRACT

Psoralen plus UVA (PUVA) is used as a very effective treatment modality for various diseases, including psoriasis and cutaneous T-cell lymphoma. PUVA-induced immune suppression and/or apoptosis are thought to be responsible for the therapeutic action. However, the molecular mechanisms by which PUVA acts are not well understood. We have previously identified platelet-activating factor (PAF), a potent phospholipid mediator, as a crucial substance triggering ultraviolet B radiation-induced immune suppression. In this study, we used PAF receptor knockout mice, a selective PAF receptor antagonist, a COX-2 inhibitor (presumably blocking downstream effects of PAF), and PAF-like molecules to test the role of PAF receptor binding in PUVA treatment. We found that activation of the PAF pathway is crucial for PUVA-induced immune suppression (as measured by suppression of delayed type hypersensitivity to Candida albicans) and that it plays a role in skin inflammation and apoptosis. Downstream of PAF, interleukin-10 was involved in PUVA-induced immune suppression but not inflammation. Better understanding of PUVA's mechanisms may offer the opportunity to dissect the therapeutic from the detrimental (ie, carcinogenic) effects and/or to develop new drugs (eg, using the PAF pathway) that act like PUVA but have fewer side effects.


Subject(s)
Apoptosis/drug effects , Apoptosis/radiation effects , Immune Tolerance/drug effects , Immune Tolerance/radiation effects , PUVA Therapy , Platelet Activating Factor/immunology , Animals , Candida albicans/immunology , Candidiasis/drug therapy , Candidiasis/immunology , Candidiasis/pathology , Combined Modality Therapy/methods , Female , Ficusin/pharmacology , Humans , Hypersensitivity, Delayed/drug therapy , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Immune Tolerance/immunology , Immunosuppression Therapy/methods , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Interleukin-10/immunology , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/immunology , Lymphoma, T-Cell, Cutaneous/pathology , Male , Mice , Mice, Knockout , PUVA Therapy/adverse effects , PUVA Therapy/methods , Platelet Membrane Glycoproteins/deficiency , Platelet Membrane Glycoproteins/immunology , Psoriasis/drug therapy , Psoriasis/immunology , Psoriasis/pathology , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Signal Transduction/radiation effects
5.
Photochem Photobiol ; 81(1): 46-51, 2005.
Article in English | MEDLINE | ID: mdl-15496135

ABSTRACT

We previously demonstrated that CD1d knockout mice were resistant to ultraviolet (UV)-induced immunosuppression. Because immune suppression is a critical factor in the development of UV-induced skin cancers, we investigated the response of wild type (WT) and CD1d-/- mice to UV carcinogenesis. We found that although 100% of WT mice developed skin tumors after 45 weeks of UV irradiation, only 60% of CD1d-/- mice developed skin tumors. To investigate the mechanisms involved in the resistance of CD1d-/- mice to UV-induced carcinogenesis, we determined the time course and kinetics of keratinocyte cell death after UV irradiation. After acute UV exposure, the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL)-positive keratinocytes were eliminated from the skin of WT mice by 72 h post-UV, but they still persisted until 96 h in CD1d-/- mice. The kinetics of p53 protein expression closely followed the kinetics of apoptotic cell death. Chronic UV irradiation resulted in induction of a significantly higher number of apoptotic keratinocytes in CD1d-/- than WT mice. In addition, epidermis and dermis from chronically UV-irradiated CD1d-/- mice harbored significantly fewer p53 mutations than WT mice. These results indicate that the resistance of CD1d-/- mice to UV carcinogenesis may be due to increased cell death and elimination of keratinocytes and fibroblasts containing DNA damage and p53 mutations.


Subject(s)
Antigens, CD1/physiology , Apoptosis , Neoplasms, Radiation-Induced/pathology , Skin Neoplasms/pathology , Ultraviolet Rays , Animals , Antigens, CD1/genetics , Antigens, CD1d , Genes, p53 , Mice , Mice, Knockout , Mutation , Neoplasms, Radiation-Induced/genetics , Skin Neoplasms/genetics
6.
Am J Pathol ; 165(3): 879-87, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331412

ABSTRACT

Inhibition of p53-induced epidermal apoptosis, generation of p53 mutations, and suppressor T cells are the critical events responsible for the induction and development of UV-induced skin cancers. Recently, we demonstrated that CD1d knockout mice were resistant to UV-induced immunosuppression, prompting us to further address the role of CD1d in regulating UV carcinogenesis. We, therefore, investigated the response of wild-type (WT) and CD1d-/- mice to UV carcinogenesis. We found that although 100% of WT mice developed skin tumors after 45 weeks of UV irradiation, only 60% of CD1d-/- mice developed skin tumors. Surprisingly, keratinocytes and fibroblasts from CD1d-/- mice were more sensitive to UV-induced apoptosis and persisted longer than cells derived from WT mice. In addition, epidermis and dermis taken from chronically UV-irradiated CD1d-/- mice harbored significantly fewer p53 mutations than WT mice. Our findings identify an unexpected and novel function for CD1d as a critical molecule regulating UV carcinogenesis, by inhibiting apoptosis to prevent elimination of potentially malignant keratinocytes and fibroblasts.


Subject(s)
Antigens, CD1/physiology , Apoptosis , Genetic Predisposition to Disease , Neoplasms, Radiation-Induced/pathology , Radiation Tolerance , Skin Neoplasms , Skin Neoplasms/pathology , Animals , Antigens, CD1/genetics , Antigens, CD1d , Apoptosis/genetics , Apoptosis/radiation effects , Female , Fibroblasts/radiation effects , Homozygote , In Situ Nick-End Labeling , Keratinocytes/radiation effects , Male , Mice , Mice, Knockout , Mutation/genetics , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/prevention & control , Skin/radiation effects , Skin Neoplasms/genetics , Skin Neoplasms/prevention & control , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
7.
Toxicol Appl Pharmacol ; 195(3): 331-8, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15020195

ABSTRACT

Applying military jet fuel (JP-8) or commercial jet fuel (Jet-A) to the skin of mice suppresses the immune response in a dose-dependent manner. The release of biological response modifiers, particularly prostaglandin E2 (PGE2), is a critical step in activating immune suppression. Previous studies have shown that injecting selective cyclooxygenase-2 inhibitors into jet fuel-treated mice blocks immune suppression. Because the inflammatory phospholipid mediator, platelet-activating factor (PAF), up-regulates cyclooxygenase-2 production and PGE2 synthesis by keratinocytes, we tested the hypothesis that PAF-receptor binding plays a role in jet fuel-induced immune suppression. Treating keratinocyte cultures with PAF and/or jet fuel (JP-8 and Jet-A) stimulates PGE2 secretion. Jet fuel-induced PGE2 production was suppressed by treating the keratinocytes with specific PAF-receptor antagonists. Injecting mice with PAF, or treating the skin of the mice with JP-8, or Jet-A, induced immune suppression. Jet fuel-induced immune suppression was blocked when the jet fuel-treated mice were injected with PAF-receptor antagonists before treatment. Jet fuel treatment has been reported to activate oxidative stress and treating the mice with anti-oxidants (Vitamins C, or E or beta-hydroxy toluene), before jet fuel application, interfered with immune suppression. These findings confirm previous studies showing that PAF-receptor binding can modulate immune function. Furthermore, they suggest that PAF-receptor binding may be an early event in the induction of immune suppression by immunotoxic environmental agents that target the skin.


Subject(s)
Hydrocarbons/toxicity , Hypersensitivity, Delayed/chemically induced , Keratinocytes/drug effects , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Administration, Cutaneous , Animals , Ascorbic Acid/therapeutic use , Cell Line , Dinoprostone/analysis , Dinoprostone/immunology , Female , Hypersensitivity, Delayed/drug therapy , Hypersensitivity, Delayed/immunology , Keratinocytes/immunology , Keratinocytes/metabolism , Mice , Platelet Activating Factor/analogs & derivatives , Platelet Activating Factor/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/immunology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/immunology , Vitamin E/therapeutic use
8.
Immunology ; 110(3): 296-303, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14632656

ABSTRACT

T-cell homing within germinal centres (GCs) is required for humoral B-cell responses. However, the mechanisms implicated in the recruitment of T cells into the GC are not completely understood. Here we show, by immunohistology, and Northern and Western blots, that in vivo human GC B lymphocytes can express CxC and CC chemokines. Moreover, B-cell subset-specific experiments reveal that interleukin (IL)-8 and regulated on activation, normal, T-cell expressed, and secreted (RANTES) are predominantly expressed by GC centroblast and centrocytes, suggesting that chemokine expression is essential at stages in which B-lymphocytes engage in active antigen-dependent interactions with T lymphocytes. In keeping with this hypothesis, we show that the T cells recruited into the GC correlatively express the receptors for IL-8 and RANTES. We propose that chemokine expression is a key B-cell function that facilitates T-lymphocyte recruitment into the GCs and supports cognate B-cell : T-cell encounters. Moreover, our data are consistent with the impaired homing of T cells to secondary lymphoid organs in mice that are either deficient in CC and CxC chemokines or their receptors.


Subject(s)
B-Lymphocyte Subsets/immunology , Chemotaxis, Leukocyte/immunology , Germinal Center/immunology , Interleukin-8/metabolism , T-Lymphocytes/immunology , Antigens, CD19/metabolism , Chemokine CCL5/immunology , Chemokine CCL5/metabolism , Humans , Lymphocyte Activation/immunology , Lymphocyte Cooperation/immunology , Receptors, CCR5/metabolism , Receptors, Interleukin-8A/metabolism
9.
J Invest Dermatol ; 119(3): 600-8, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12230501

ABSTRACT

The ultraviolet radiation present in sunlight is immune suppressive. Recently we showed that solar-simulated ultraviolet radiation (ultraviolet A + B; 295-400 nm), applied after immunization, suppressed immunologic memory and the elicitation of delayed-type hypersensitivity to the common opportunistic pathogen, Candida albicans. Further, we found that wavelengths in the ultraviolet A region of the solar spectrum (320-400 nm), devoid of ultraviolet B, were equally effective in activating immune suppression as ultraviolet A + B radiation. Here we report on the mechanisms involved. Maximal immune suppression was found when mice were exposed to solar-simulated ultraviolet radiation 7-9 d post immunization. No immune suppression was found in ultraviolet-irradiated mice injected with monoclonal anti-interleukin-10 antibody, or mice exposed to solar-simulated ultraviolet radiation and injected with recombinant interleukin-12. Suppressor lymphocytes were found in the spleens of mice exposed to ultraviolet A + B radiation. In addition, antigen-specific suppressor T cells (CD3+, CD4+, DX5+) were found in the spleens of mice exposed to ultraviolet A radiation. Applying liposomes containing bacteriophage T4N5 to the skin of mice exposed to solar-simulated ultraviolet A + B radiation, or mice exposed to ultraviolet A radiation, blocked immune suppression, demonstrating an essential role for ultraviolet-induced DNA damage in the suppression of established immune reactions. These findings indicate that overlapping immune suppressive mechanisms are activated by ultraviolet A and ultraviolet A + B radiation. Moreover, our findings demonstrate that ultraviolet radiation activates similar immunologic pathways to suppress the induction of, or the elicitation of, the immune response.


Subject(s)
Immune System/immunology , Immune System/radiation effects , Ultraviolet Rays/adverse effects , Animals , Antibodies, Monoclonal/pharmacology , DNA Damage/immunology , DNA Repair/immunology , Female , Hypersensitivity, Delayed/immunology , Immune System/drug effects , Interleukin-10/immunology , Interleukin-10/pharmacology , Interleukin-12/immunology , Interleukin-12/pharmacology , Mice , Mice, Inbred C3H , Neoplasms, Radiation-Induced/immunology , Skin Neoplasms/immunology , Specific Pathogen-Free Organisms , Sunlight/adverse effects , T-Lymphocytes/immunology , T-Lymphocytes/radiation effects
10.
Methods ; 28(1): 25-33, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12231185

ABSTRACT

Ultraviolet (UV) radiation present in sunlight plays a critical role in the initiation and promotion of nonmelanoma skin carcinogenesis and immune suppression. The immune suppressive effects of UV have been identified as a risk factor for skin cancer induction. For these reasons, scientists have focused on elucidating the mechanisms of UV-induced immune suppression to better understand the pathogenesis of skin cancer induction. A hallmark of UV-induced immune suppression is the generation of antigen-specific suppressor T cells. These suppressor cells have been shown to suppress antitumor immunity as well as other cell-mediated responses such as delayed-type hypersensitivity (DTH) reactions. Due to the excessive cost and time involved in traditional UV carcinogenic experiments, scientists have opted to use UV-induced suppression of DTH reactions as a surrogate model. DTH has been, and continues to be, a widely used assay system to measure in vivo immune function. Although somewhat unsophisticated by today's standards, this assay has great advantages because it presents a fast, inexpensive, and reliable model system to help dissect the mechanisms involved in UV-induced immune suppression. Furthermore, the murine model of DTH enables scientists to perform additional procedures, such as adoptive transfer studies with suppressor T cells, which are currently unavailable with human subjects.


Subject(s)
Hypersensitivity, Delayed/immunology , Immune Tolerance/radiation effects , Ultraviolet Rays , Adoptive Transfer , Animals , Female , Mice , Mice, Inbred Strains
11.
Methods ; 28(1): 71-8, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12231190

ABSTRACT

Ultraviolet radiation exposure damages DNA and promotes the development of skin cancer. In addition, UV exposure suppresses the immune response. Although the mechanism by which epidermal exposure to UV induces systemic immune suppression is not fully understood, it is clear that cytokines are involved. Therefore, quantitative measurement of cytokines is a critical aspect of modern research techniques. Determining the level of synthesis and secretion of cytokines in vivo or in vitro can be achieved through several possible techniques, depending on the sampling size, its physical state, and the type of answers required to test the hypothesis. When studying transcriptional activation, the level of cytokine mRNA is often determined using reverse transcription polymerase chain reaction (RT-PCR), ribonuclease protection assay (RPA), or Northern blot. Quantitative determinations of specific protein levels require a capture ELISA. As with any analytical technique, there are compromises among expense of sensitivity, labor, and time. These methods are discussed as they pertain to surveying cytokine induction and their relative usefulness to the laboratory scientist.


Subject(s)
Cytokines/physiology , Immune Tolerance/radiation effects , Ultraviolet Rays , Animals , Cytokines/analysis , Cytokines/genetics , Enzyme-Linked Immunosorbent Assay , Humans , Reverse Transcriptase Polymerase Chain Reaction
12.
Toxicol Appl Pharmacol ; 180(2): 136-44, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11969381

ABSTRACT

Applying military jet fuel (JP-8) to the skin of mice activates systemic immune suppression. In all of our previous experiments, JP-8 was applied to immunologically naïve mice. The effect of jet fuels on established immune reactions, such as immunological memory, is unknown. The focus of the experiments presented here was to test the hypothesis that jet fuel exposure [both JP-8 and commercial jet fuel (Jet-A)] suppresses established immune reactions. Mice were immunized with the opportunistic fungal pathogen Candida albicans and, at different times after immunization (10 to 30 days), various doses of undiluted JP-8 or Jet-A were applied to their skin. Both the elicitation of delayed-type hypersensitivity (DTH) (mice challenged 10 days after immunization) and immunological memory (mice challenged 30 days after immunization) were significantly suppressed in a dose-dependent manner. Dermal exposure to either multiple small doses (50 microl over 4 days) or a single large dose (approximately 200-300 microl) of JP-8 and/or Jet-A suppressed DTH to C. albicans. The mechanism by which dermal application of JP-8 and Jet-A suppresses immunological memory involves the release of immune biologic response modifiers. Blocking the production of prostaglandin E(2) by a selective cyclooxygenase-2 inhibitor (SC 236) significantly reversed jet fuel-induced suppression of immunologic memory. These findings indicate, for the first time, that dermal exposure to commercial jet fuel (Jet-A) suppresses the immune response. In addition, the data reported here expand on previous findings by suggesting that jet fuel exposure may depress the protective effect of prior vaccination.


Subject(s)
Hydrocarbons/toxicity , Hypersensitivity, Delayed/chemically induced , Immunologic Memory/immunology , Animals , Candida albicans/immunology , Cyclooxygenase Inhibitors/pharmacology , Female , Hydrocarbons/immunology , Hypersensitivity, Delayed/immunology , Immunization , Immunologic Memory/drug effects , Mice , Mice, Inbred C3H , Pyrazoles/pharmacology , Skin Absorption/immunology , Specific Pathogen-Free Organisms , Sulfonamides/pharmacology
13.
J Exp Med ; 195(2): 171-9, 2002 Jan 21.
Article in English | MEDLINE | ID: mdl-11805144

ABSTRACT

Ultraviolet (UV) radiation plays a critical role in the induction of nonmelanoma skin cancer. UV radiation is also immune suppressive, and the immune suppression induced by UV irradiation has been identified as a major risk factor for skin cancer induction. Previously, we showed that UV exposure activates a cytokine cascade involving prostaglandin (PG)E(2), interleukin (IL)-4, and IL-10 that induces immune suppression. However, the earliest molecular events that occur immediately after UV exposure, especially those upstream of PGE2, are not well defined. UV-irradiated keratinocytes secrete the inflammatory phospholipid mediator, platelet-activating factor (PAF). Because PAF upregulates the production of immunomodulatory compounds, including PGE2, we tested the hypothesis that UV-induced PAF activates cytokine production and initiates UV-induced immune suppression. Both UV and PAF activated cyclooxygenase (COX)-2 and IL-10 reporter gene construct transcription. PAF mimicked the effects of UV in vivo and suppressed delayed-type hypersensitivity (DTH). Furthermore, immune suppression was blocked when UV-irradiated mice were injected with PAF receptor antagonists. In addition to the well-known role of PAF as a proinflammatory lipid mediator, we propose that the PAF receptor senses cellular damage through the recognition of PAF and/or PAF-like molecules, such as oxidized phosphatidylcholine, which activates cytokine transcription and induces systemic immune suppression.


Subject(s)
Immunosuppression Therapy , Keratinocytes/immunology , Platelet Activating Factor/immunology , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Animals , Cyclooxygenase 2 , Enzyme Activation/radiation effects , Isoenzymes/immunology , Isoenzymes/metabolism , Keratinocytes/metabolism , Keratinocytes/radiation effects , Mice , Mice, Inbred C3H , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/immunology , Platelet Membrane Glycoproteins/metabolism , Prostaglandin-Endoperoxide Synthases/immunology , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Interleukin/biosynthesis , Receptors, Interleukin/immunology , Receptors, Interleukin-10 , Signal Transduction/immunology , Transcription, Genetic/radiation effects , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...