Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 216
Filter
1.
J Invest Dermatol ; 117(5): 1193-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11710932

ABSTRACT

The ultraviolet radiation present in sunlight is the primary cause of nonmelanoma skin cancer and has been implicated in the development of cutaneous malignant melanoma. In addition, ultraviolet is immune suppressive and the suppression induced by ultraviolet radiation has been identified as a risk factor for skin cancer induction. Ultraviolet also suppresses the immune response to infectious agents. In most experimental models, ultraviolet is applied to immunologically naive animals prior to immunization. Of equal concern, however, is the ability of sunlight to suppress established immune reactions, such as the recall reaction in humans, which protects against microbial infections. Here we demonstrate that solar-simulated ultraviolet radiation, applied after immunization, suppresses immunologic memory and the elicitation of delayed-type hypersensitivity. Further, we found that wavelengths in the ultraviolet A region of the solar spectrum were critical for inducing immune suppression. Ultraviolet A (320-400 nm) radiation was as effective as solar-simulated ultraviolet A + B (290-400 nm) in suppressing the elicitation of an established immune response. Irradiation with ultraviolet AI (340-400 nm) had no effect. Supporting a critical role for ultraviolet A in ultraviolet-induced immune suppression was the observation that applying a sunscreen that contained an ultraviolet B only filter had no protective effect, whereas, a sunscreen containing both ultraviolet A and ultraviolet B filters totally blocked ultraviolet-induced immune suppression. These data suggest that sunlight may depress the protective effect of prior vaccination. In addition, the observation that ultraviolet A is immunosuppressive indicates the need for ultraviolet A protection when designing sun protection strategies.


Subject(s)
Immune System/radiation effects , Ultraviolet Rays , Animals , Antibody Formation/drug effects , Antibody Formation/radiation effects , Chemistry, Pharmaceutical , Hypersensitivity, Delayed/prevention & control , Immune System/drug effects , Immunization , Immunologic Memory/radiation effects , Mice , Mice, Inbred C3H , Sunscreening Agents/chemistry , Sunscreening Agents/pharmacology
2.
Proc Natl Acad Sci U S A ; 98(17): 9790-5, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11481437

ABSTRACT

Epidemiological studies have suggested an association between exposure to solar UV radiation and the incidence of lymphoid malignancies, which has increased substantially worldwide during the last two decades. Findings from animal studies have raised the question of whether UV radiation might influence the development of lymphoid malignancies by means of its immunosuppressive effect. In this study, we examined the effect of UV irradiation on the development of lymphoid malignancies in mice with no or only one functional copy of p53. Mice that lack both copies of p53 spontaneously develop high frequency of lymphoid malignancies in the thymus and spleen. p53 heterozygous mice with only one copy of the wild-type allele also develop lymphoid malignancies, but with a much lower frequency and a long latent period. In our study using mice of the C57BL/6 background, only one of the unirradiated mice lacking one copy of p53 (p53(+/-)) spontaneously developed a lymphoid tumor (6%), whereas 88% of UV-irradiated p53(+/-) mice developed lymphoid tumors in the spleen or liver. None of the control or UV-irradiated p53 wild-type mice developed lymphoid tumors during the 60-week observation period. Both UV-irradiated and unirradiated mice lacking both copies of p53 (p53(-/-)) rapidly developed thymic lymphomas and/or lymphoid tumors in spleen or liver. All of the lymphoid tumors tested were of T cell type. The immune responses of the mice to contact sensitization were identical and were suppressed to the same extent by UV irradiation regardless of the genotype. These results indicate that differences in immune reactivity do not account for the different effects of UV radiation on lymphoid malignancies and, in addition, that p53 is not required for generation of T cell-mediated immunity. Interestingly, whereas p53 mutations or loss of heterozygosity did not account for the accelerated development of lymphoid tumors in UV-irradiated p53(+/-) mice, deletions in the p16(INK4a) gene were quite common. These data provide the experimental evidence that UV irradiation induces lymphoid neoplasms in genetically susceptible mice and support the hypothesis that extensive sunlight exposure contributes to the induction of lymphoma in humans.


Subject(s)
Gene Deletion , Genes, p53 , Lymphoma, Non-Hodgkin/etiology , Neoplasms, Radiation-Induced/genetics , Ultraviolet Rays/adverse effects , Age Factors , Animals , Animals, Congenic , Genes, p16/radiation effects , Hypersensitivity, Delayed/genetics , Hypersensitivity, Delayed/immunology , Immunity, Cellular/genetics , Immunity, Cellular/radiation effects , Liver Neoplasms/etiology , Liver Neoplasms/genetics , Lymphoma, Non-Hodgkin/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Specific Pathogen-Free Organisms , Splenic Neoplasms/etiology , Splenic Neoplasms/genetics , Thymus Neoplasms/etiology , Thymus Neoplasms/genetics
3.
Photochem Photobiol ; 73(5): 537-44, 2001 May.
Article in English | MEDLINE | ID: mdl-11367577

ABSTRACT

These studies addressed the hypothesis that UV radiation (UVR) could affect immune responses in mice infected with Borrelia burgdorferi. Immunity against the Lyme spirochete B. burgdorferi was studied in a murine model of UV-induced immune suppression. Borrelia-specific cellular and humoral responses were examined following immunosuppressive doses of UVR. Low-passage Borrelia were injected intradermally at the base of the tail following irradiation. At various time points after infection the blood was cultured for the presence of Borrelia and the serum analyzed for Borrelia-specific antibodies. Two weeks after infection one hind-limb joint was cultured for the presence of spirochetes and the contralateral joint was examined histologically for arthritis formation. The results demonstrated that UV irradiation, administered at the site of infection or at a distant site, suppressed Borrelia-specific cellular and humoral responses in infected mice. Suppression of delayed-type hypersensitivity and antibody responses to UV was abrogated by administration of anti-interleukin (IL)-10 after UV irradiation. In addition, UV irradiation altered the dissemination pattern of the bacteria from the skin into the blood and exacerbated arthritis when compared with unirradiated controls. From these studies we concluded that UV irradiation can modulate the immune response to Borrelia and exacerbate the subsequent arthritic component of Lyme disease in mice. Furthermore, our studies suggest that IL-10 is in part responsible for the suppression of both cellular and humoral responses in addition to playing a role in the development of Lyme arthritis.


Subject(s)
Borrelia burgdorferi Group/immunology , Lyme Disease/immunology , Ultraviolet Rays , Adoptive Transfer , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Female , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/microbiology , Interleukin-10/immunology , Mice , Mice, Inbred C3H , Th1 Cells/immunology
4.
Cancer Res ; 61(1): 215-21, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11196164

ABSTRACT

We have developed a murine melanoma model that allows us to investigate the mechanisms by which spontaneous, immunogenic melanoma metastases escape immunological destruction in syngeneic mice. In the current study, we tested the hypothesis that loss of immunogenicity is an obligatory step in the persistence of pulmonary metastases. Fragments of syngeneic K1735-M2 tumor were implanted in the outer edge of one pinna per C3H/HeN mouse, and the growing tumors were removed 2-3 weeks later. Two weeks after removal of the tumors, the mice demonstrated effective T-cell-mediated immunity to s.c. challenge with K1735-M2 cells. However, lung metastases appeared in 23% of the immunized mice within 9-12 weeks after the initial tumor implantation. The expression of protective immunity to s.c. tumors required the presence of both CD4+ and CD8+ T cells. The immunized mice had specific CTLs capable of killing both K1735-M2 melanoma cells and the cells of nine independently derived melanoma metastases. Furthermore, K1735-M2 immunization protected these mice from s.c. tumor challenge with all nine metastatic cell lines. Our results demonstrate that the persistence of these metastases within the lung was not attributable to emergence of antigen-loss variants in immunized hosts. Our model provides an approach to investigate other mechanisms by which spontaneous metastases escape from immunological control and an opportunity to improve immunotherapy of melanoma metastases.


Subject(s)
Lung Neoplasms/immunology , Lung Neoplasms/secondary , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Animals , Antigens, Neoplasm/immunology , Cytotoxicity, Immunologic , Disease Models, Animal , Hypersensitivity, Delayed/immunology , Immunity, Innate/immunology , Immunization , Male , Mice , Mice, Inbred C3H , Neoplasm Transplantation , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology
5.
Photochem Photobiol ; 71(6): 743-6, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10857371

ABSTRACT

The objective of this study was to determine whether exposure of early suckling young of the opossum Monodelphis domestica to ultraviolet A (UVA) radiation (320-400 nm) can lead to the development of melanocytic lesions similar to those induced after exposure to ultraviolet B (UVB) radiation (280-320 nm) to total doses as low as 380 J/m2. A total of 576 sucklings received nine exposures of 0.6, 2.6 or 15.5 kJ/m2 per dose (total doses approximately 6, 23 and 140 kJ/m2, respectively) from a Blak Ray lamp source with a narrow range emission at 365 nm. A further 280 sucklings were exposed in the same way to doses of 2.6 kJ/m2 per dose (total approximately 23 kJ/m2) broad-band UVA with visible wavelengths from a Dermalight lamp. Frequency of litter loss following all of the UVA-exposure protocols was similar to that within the same stocks in the colony at large. Only one of the 856 UVA-exposed individuals possessed a melanocytic lesion at the 5 month assessment point. No radiation-induced lesions of any type were evident on the skin of the other animals exposed as sucklings. The affected male was from a group of 70 individuals exposed to the highest total dose (140 kJ/m2) from the Blak Ray light source. The melanocytic hyperplasia was provisionally identified as a potential melanoma but it slowly regressed as the animal aged. We conclude that in the opossum suckling exposure system, the potency of UVA for melanoma induction is extremely low compared with that of UVB. Possible explanations, amenable to further investigations, are given for the low UVA sensitivity of the suckling model compared to the adult exposure model of Ley (Ley, R. D. [1997] Cancer Res. 57, 3682-3684).


Subject(s)
Animals, Suckling , Melanoma, Experimental/pathology , Neoplasms, Radiation-Induced/pathology , Radiation Dosage , Skin Neoplasms/pathology , Animals , Disease Models, Animal , Male , Opossums , Ultraviolet Rays
6.
J Exp Med ; 191(11): 2011-20, 2000 Jun 05.
Article in English | MEDLINE | ID: mdl-10839815

ABSTRACT

Whereas CD40-CD40 ligand interactions are important for various dendritic cell (DC) functions in vitro, their in vivo relevance is unknown. We analyzed the DC status of CD40 ligand -/- mice using a contact hypersensitivity (CHS) model system that enables multiple functions of DCs to be assessed in vivo. Immunohistochemistry of skin sections revealed no differences in terms of numbers and morphology of dendritic epidermal Langerhans cells (LCs) in unsensitized CD40 ligand -/- mice as compared with wild-type C57BL/6 mice. However, after contact sensitization of CD40 ligand -/- mice, LCs failed to migrate out of the skin and substantially fewer DCs accumulated in draining lymph nodes (DLNs). Furthermore, very few antigen-bearing DCs could be detected in the paracortical region of lymph nodes draining sensitized skin. This defect in DC migration after hapten sensitization was associated with defective CHS responses and decreased cutaneous tumor necrosis factor (TNF)-alpha production and was corrected by injecting recombinant TNF-alpha or an agonistic anti-CD40 monoclonal antibody. Thus, CD40-CD40 ligand interactions in vivo regulate the migration of antigen-bearing DCs from the skin to DLNs via TNF-alpha production and play a vital role in the initiation of acquired T cell-mediated immunity.


Subject(s)
CD40 Antigens/immunology , Cell Movement/immunology , Langerhans Cells/immunology , Membrane Glycoproteins/immunology , Animals , CD40 Ligand , Cell Count , Dermatitis, Contact , Disease Models, Animal , Langerhans Cells/classification , Langerhans Cells/cytology , Langerhans Cells/physiology , Lymph Nodes , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/cytology , Tumor Necrosis Factor-alpha/biosynthesis
7.
J Invest Dermatol ; 114(1): 149-56, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10620131

ABSTRACT

Exposing human skin to ultraviolet radiation causes DNA damage, sunburn, immune alterations, and eventually, skin cancer. We wished to determine whether liposomes containing a DNA repair enzyme could prevent any of the acute effects of irradiation when applied after ultraviolet exposure. Fifteen human patients with a prior history of skin cancer were exposed to two minimal erythema doses of ultraviolet radiation on their buttock skin. Liposomes containing T4 endonuclease V or heat-inactivated enzyme were applied immediately and at 2, 4, and 5 h after ultraviolet irradiation. Transmission electron microscopy after anti-T4 endonuclease V-staining and immunogold labeling on biopsies taken at 6 h after ultraviolet exposure revealed that the enzyme was present within cells in the skin. Immunohistochemical DNA damage studies suggested a trend toward improved DNA repair at the active T4 endonuclease V liposome-treated test sites. Although the active T4 endonuclease V liposomes did not significantly affect the ultraviolet-induced erythema response and microscopic sunburn cell formation, they nearly completely prevented ultraviolet-induced upregulation of interleukin-10 and tumor necrosis factor-alpha RNA message and of interleukin-10 protein. These studies demonstrate that liposomes can be used for topical intracellular delivery of small proteins to human skin and suggest that liposomes containing DNA repair enzymes may provide a new avenue for photoprotection against some forms of ultraviolet-induced skin damage.


Subject(s)
DNA Ligases/administration & dosage , Endodeoxyribonucleases/administration & dosage , Interleukin-10/metabolism , Radiation-Protective Agents/administration & dosage , Skin/drug effects , Skin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ultraviolet Rays , Viral Proteins , Administration, Topical , Adult , Aged , DNA Ligases/pharmacokinetics , DNA Ligases/pharmacology , DNA Repair/drug effects , Deoxyribonuclease (Pyrimidine Dimer) , Drug Carriers , Endodeoxyribonucleases/pharmacokinetics , Endodeoxyribonucleases/pharmacology , Female , Humans , Keratinocytes/enzymology , Langerhans Cells/enzymology , Liposomes , Male , Microscopy, Electron , Middle Aged , Radiation-Protective Agents/pharmacokinetics , Radiation-Protective Agents/pharmacology , Skin/radiation effects , Skin/ultrastructure , Time Factors , Up-Regulation/drug effects
8.
Immunity ; 11(5): 625-36, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10591187

ABSTRACT

We examined dendritic cell (DC) status in SCID and RAG2 -/- mice to assess the influence of T cells on DC development and function in vivo. These mice have reduced numbers of DC in the epidermis and lymph nodes draining hapten-sensitized skin. Epidermal DC in these mice were defective in presenting antigen in vivo to adoptively transferred, hapten-sensitized T cells from normal mice. Likewise, draining lymph node DC were deficient in their capacity to stimulate naive T cells in vitro and in vivo. DC numbers as well as the impaired ability to present antigen in vivo, were corrected by reconstituting these animals with normal T lymphocytes, suggesting that T cells are crucial for normal DC maturation and function in vivo.


Subject(s)
Dendritic Cells/cytology , T-Lymphocyte Subsets/physiology , Adoptive Transfer , Animals , Antigen Presentation , Cell Count , Cell Differentiation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/pathology , Epidermis/immunology , Epidermis/pathology , Haptens/immunology , Langerhans Cells/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , T-Lymphocyte Subsets/transplantation
9.
J Investig Dermatol Symp Proc ; 4(1): 65-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10537011

ABSTRACT

In order to protect the public against the adverse effects of sunlight, the scientific, medical, and particularly the dermatologic community has promoted "safe sun exposure." This strategy includes sun avoidance whenever possible, wearing hats and other protective clothing and/or devices, such as sunglasses, and extensive use of sunscreens. Sunscreen efficacy is determined by measuring the ability of the sunscreen to block ultraviolet (UV)-induced erythema (sun protection factor or SPF), and most sunscreen formulations on the market, if used properly, are very good at preventing erythema and sunburn. How well sunscreens protect against the other adverse effects of sunlight, such as immune suppression, is not as clear. The purpose of this paper is to review and discuss the literature in this area, concentrating on some of the complications of determining how well sunscreens protect against UV-induced immune suppression.


Subject(s)
Immune System/radiation effects , Skin/immunology , Sunscreening Agents/pharmacology , Ultraviolet Rays , Animals , Humans , Hypersensitivity, Delayed/immunology , Immune System/drug effects , Protective Devices , Skin/drug effects
10.
Science ; 285(5429): 898-900, 1999 Aug 06.
Article in English | MEDLINE | ID: mdl-10436160

ABSTRACT

DNA-damaged cells can either repair the DNA or be eliminated through a homeostatic control mechanism termed "cellular proofreading." Elimination of DNA-damaged cells after ultraviolet radiation (UVR) through sunburn cell (apoptotic keratinocyte) formation is thought to be pivotal for the removal of precancerous skin cells. Sunburn cell formation was found to be dependent on Fas ligand (FasL), a pro-apoptotic protein induced by DNA damage. Chronic exposure to UVR caused 14 of 20 (70 percent) FasL-deficient mice and 1 of 20 (5 percent) wild-type mice to accumulate p53 mutations in the epidermis. Thus, FasL-mediated apoptosis is important for skin homeostasis, suggesting that the dysregulation of Fas-FasL interactions may be central to the development of skin cancer.


Subject(s)
DNA Damage , Epidermal Cells , Genes, p53 , Keratinocytes/cytology , Membrane Glycoproteins/physiology , Skin Neoplasms/etiology , Animals , Apoptosis , Epidermis/metabolism , Epidermis/radiation effects , Fas Ligand Protein , Keratinocytes/metabolism , Keratinocytes/radiation effects , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C3H , Mutation , Skin Neoplasms/pathology , Ultraviolet Rays , Up-Regulation , fas Receptor/genetics , fas Receptor/physiology
11.
Oncogene ; 18(29): 4247-53, 1999 Jul 22.
Article in English | MEDLINE | ID: mdl-10435637

ABSTRACT

To assess the role of the p53 tumor suppressor gene in skin carcinogenesis by UV radiation, mice constitutively lacking one or both copies of the functional p53 gene were compared to wild-type mice for their susceptibility to UV carcinogenesis. Heterozygous mice showed greatly increased susceptibility to skin cancer induction, and homozygous p53 knockout mice were even more susceptible. Accelerated tumor development in the heterozygotes was not associated with loss of the remaining wild-type allele of p53, as reported for tumors induced by other carcinogens, but in many cases was associated with UV-induced mutations in p53. Tumors arose on the ears and dorsal skin of mice of all three genotypes, and homozygous knockout mice also developed ocular tumors, mainly melanomas. Skin tumors in the p53 knockout mice were predominately squamous cell carcinomas and were associated with premalignant lesions resembling actinic keratoses, whereas those in the heterozygous and wild-type mice were mainly sarcomas. These results demonstrate the importance of p53 in protecting against UV-induced cancers, particularly in the eye and epidermis.


Subject(s)
Carcinoma, Squamous Cell/prevention & control , Epidermis/radiation effects , Eye Neoplasms/prevention & control , Genes, p53 , Melanoma, Experimental/prevention & control , Neoplasms, Radiation-Induced/prevention & control , Radiation Tolerance/genetics , Sarcoma, Experimental/prevention & control , Skin Neoplasms/prevention & control , Tumor Suppressor Protein p53/physiology , Ultraviolet Rays/adverse effects , Animals , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/genetics , Codon/genetics , Crosses, Genetic , DNA, Neoplasm/genetics , Epidermis/metabolism , Exons/genetics , Eye Neoplasms/etiology , Eye Neoplasms/genetics , Gene Dosage , Genetic Predisposition to Disease , Genotype , Keratosis/etiology , Keratosis/genetics , Melanoma, Experimental/etiology , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms, Radiation-Induced/genetics , Point Mutation , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Precancerous Conditions/etiology , Precancerous Conditions/genetics , Sarcoma, Experimental/etiology , Sarcoma, Experimental/genetics , Skin Neoplasms/etiology , Skin Neoplasms/genetics , Specific Pathogen-Free Organisms , Tumor Suppressor Protein p53/deficiency
12.
Photochem Photobiol ; 69(5): 571-4, 1999 May.
Article in English | MEDLINE | ID: mdl-10333763

ABSTRACT

Exposure to sunlight can result in a number of harmful effects, including sunburn, erythema, premature aging of the skin, immune suppression and skin cancer. Studies designed to understand the underlying mechanisms often depend upon the use of artificial sources of UV radiation. Unfortunately, conclusions from different laboratories using different lamps often conflict, and it is entirely possible that the different spectra of sunlights used in each may be a source of conflict. To minimize confounding variables, we employed two of the more commonly used UV light sources, fluorescent sunlamps, such as the FS-40 and Kodacel-filtered FS-40 sunlamps, and a xenon arc solar simulator and compared, in one series of standardized experiments, the effects of each light source on DNA damage, urocanic acid isomerization and edema formation. The dose-response curves, calculated by linear regression or curve fitting were compared. The data indicate that DNA damage and urocanic acid isomerization were more sensitive to shorter wavelengths of UV than longer wavelengths, and the biological endpoint of edema most closely correlated with the induction of DNA damage. The results emphasize the dominance of shorter wavelengths within the UV spectrum in damaging biological tissues, even when the solar simulator, which contains significant amounts of UVA, was used and demonstrate that each light source has a characteristic pattern of induction of biochemical and biological endpoints.


Subject(s)
DNA Damage , Edema/etiology , Ultraviolet Rays/adverse effects , Urocanic Acid/chemistry , Animals , Female , Isomerism , Mice , Mice, Inbred C3H , Skin/pathology , Skin/radiation effects
13.
J Invest Dermatol ; 112(5): 763-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10233769

ABSTRACT

We demonstrated previously that p53 mutations can be detected in ultraviolet B-irradiated mouse skin months before the gross appearance of skin tumors and that applying sun protection factor 15 sunscreens to mouse skin before each Kodacel-filtered FS40 sunlamp irradiation resulted in the reduction of such mutations. To determine whether there is an association between reduction of ultraviolet-induced p53 mutations by sunscreens and protection against skin cancer using an environmentally relevant light source, we applied sunscreens (sun protection factors 15-22) on to the shaved dorsal skin of C3H mice 30 min before each exposure to 4.54 kJ ultraviolet B (290-400 nm) radiation per m2 from a solar simulator. Control mice were treated 5 d per wk with ultraviolet only or vehicle plus ultraviolet. p53 mutation analysis indicated that mice exposed to ultraviolet only or vehicle plus ultraviolet for 16 wk (cumulative exposure to 359 kJ ultraviolet B per m2) developed p53 mutations at a frequency of 56%-69%, respectively, but less than 5% of mice treated with sunscreens plus ultraviolet showed evidence of p53 mutations. More importantly, 100% of mice that received a cumulative dose of 1000 kJ ultraviolet B per m2 only, or vehicle plus ultraviolet B developed skin tumors, whereas, the probability of tumor development in all the mice treated with the sunscreens plus 1000 kJ ultraviolet B per m2 was 2% and mice treated with sunscreens plus 1500 kJ ultraviolet B per m2 was 15%. These results demonstrate that the sunscreens used in this study not only protect mice against ultraviolet-induced p53 mutations, but also against skin cancers induced with solar-simulated ultraviolet. Because of this association, we conclude that inhibition of p53 mutations is a useful early biologic endpoint of photoprotection against an important initiating event in ultraviolet carcinogenesis.


Subject(s)
Skin Neoplasms/genetics , Skin Neoplasms/prevention & control , Skin/drug effects , Sunscreening Agents/pharmacology , Tumor Suppressor Protein p53/genetics , Animals , Dose-Response Relationship, Radiation , Female , Mice , Mice, Inbred C3H , Mutation , Skin/radiation effects , Skin Neoplasms/epidemiology , Specific Pathogen-Free Organisms , Ultraviolet Rays
14.
J Exp Med ; 189(8): 1285-94, 1999 Apr 19.
Article in English | MEDLINE | ID: mdl-10209045

ABSTRACT

Induction of antigen-specific suppression elicited by environmental insults, such as ultraviolet (UV)-B radiation in sunlight, can inhibit an effective immune response in vivo and may contribute to the outgrowth of UV-induced skin cancer. Although UV-induced DNA damage is known to be an initiating event in the immune suppression of most antigen responses, the underlying mechanism(s) of such suppression remain undefined. In this report, we document that Fas ligand (FasL) is critical for UV-induced systemic immune suppression. Normal mice acutely exposed to UV exhibit a profound suppression of both contact hypersensitivity and delayed type hypersensitivity (DTH) reactions and the development of transferable antigen-specific suppressor cells. FasL-deficient mice exposed to UV lack both transferable suppressor cell activity and primary suppression to all antigens tested, with the exception of the DTH response to allogeneic spleen cells. Interestingly, suppression of this response is also known to occur independently of UV-induced DNA damage. Delivery of alloantigen as protein, rather than intact cells, restored the requirement for FasL in UV-induced immune suppression of this response. These results substantiate that FasL/Fas interactions are essential for systemic UV-induced suppression of immune responses that involve host antigen presentation and suggest an interrelationship between UV-induced DNA damage and FasL in this phenomenon. Collectively, our results suggest a model whereby UV-induced DNA damage disarms the immune system in a manner similar to that observed in immunologically privileged sites.


Subject(s)
Immunity/radiation effects , Membrane Glycoproteins/immunology , Animals , Antigen Presentation/immunology , Candida albicans/immunology , DNA Damage/immunology , Dermatitis, Contact/immunology , Fas Ligand Protein , Fluorescein-5-isothiocyanate , Hypersensitivity, Delayed/immunology , Isoantigens/immunology , Mice , Mice, Inbred Strains , Spleen/immunology , T-Lymphocytes/immunology , Ultraviolet Rays , fas Receptor/immunology
15.
Photochem Photobiol ; 68(5): 738-44, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9825703

ABSTRACT

Although acute exposure to UV radiation suppresses the induction of delayed-type (DTH) and contact (CHS) hypersensitivity in mice, it is not clear whether the photobiological mechanism(s) involved in suppressing these closely related immune reactions is the same. A careful examination of the UV dose responses and wavelength dependencies involved in suppressing CHS and DTH may provide important insights into the mechanisms involved. We compared the UV dose-response curves for suppressing four closely related immune reactions, local and systemic suppression of CHS to dinitrofluorobenzene, systemic suppression of DTH to Candida albicans and systemic suppression of DTH to alloantigen using three different UV spectra (FS40 sunlamps, Kodacel-filtered FS40 sunlamps and solar-simulated light). For each immune response studied, the amount of UVB radiation required to induce 50% immune suppression was lowest when FS40 sunlamps were used, highest with solar-simulated light and intermediate when Kodacel-filtered FS40 sunlamps were used, but the differences observed were not statistically significant. The UV dose-response curves for immune suppression differed significantly depending on the assay used, the site of antigenic sensitization and the antigen used. These findings suggest that the mechanisms by which UV radiation induces immune suppression differ for the four immunological reactions studied.


Subject(s)
Dermatitis, Contact/physiopathology , Hypersensitivity, Delayed/physiopathology , Skin/radiation effects , Ultraviolet Rays , Animals , Candida albicans/immunology , Dinitrofluorobenzene , Female , Isoantigens , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Skin/immunology
16.
J Invest Dermatol ; 111(5): 864-72, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9804351

ABSTRACT

We determined whether cutaneous angiogenesis induced by exposure of mice to ultraviolet-B (UVB) radiation is associated with an imbalance between positive and negative angiogenesis-regulating molecules. Unshaved C3H/HeN mice were exposed to a single dose (15 kJ per m2) of UVB. At various times, the mice were killed, and their external ears were processed for routine histology and immunohistochemistry. Antibodies against proliferating cell nuclear antigen and bromodeoxyuridine identified dividing cells. Antibodies against CD31/ PECAM-1 identified endothelial cells, and antibodies against basic fibroblast growth factor (bFGF), vascular endothelial growth factor/vascular permeability factor, and interferon-beta (IFN-beta) identified angiogenesis-regulating molecules. Epidermal hyperplasia was documented by 48 h and reached a maximum on day 7 after exposure to UVB. The expression of bFGF increased by 24 h, whereas the expression of IFN-beta decreased by 72 h after exposure to UVB. The expression of vascular endothelial growth factor/vascular permeability factor increased slightly after irradiation. The altered balance between bFGF and IFN-beta was associated with increased endothelial cell proliferation (bromodeoxyuridine + CD31 + cells) within existing blood vessels, leading to telangiectasia and new blood vessels. UV-induced epidermal hyperplasia and cutaneous angiogenesis were highest in IFN-alpha/beta receptor knockout mice. These results demonstrate that in response to UVB radiation, dividing keratinocytes produce a positive angiogenic molecule (bFGF) but not a negative angiogenic molecule (IFN-beta), and that this altered balance is associated with enhanced cutaneous angiogenesis.


Subject(s)
Neovascularization, Pathologic/etiology , Skin/blood supply , Ultraviolet Rays , Animals , Cell Division/radiation effects , Endothelial Growth Factors/analysis , Female , Fibroblast Growth Factor 2/analysis , Hyperplasia/etiology , Hyperplasia/metabolism , Interferon-alpha/analysis , Interferon-beta/analysis , Keratinocytes/cytology , Kinetics , Lymphokines/analysis , Mice , Mice, Inbred C3H , Neovascularization, Pathologic/metabolism , Skin/pathology , Skin/radiation effects , Staining and Labeling , Time Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
J Photochem Photobiol B ; 44(2): 107-11, 1998 Jul 10.
Article in English | MEDLINE | ID: mdl-9757591

ABSTRACT

Experimental data are reviewed that lend support to the hypothesis that formation of DNA damage is the initiation event of local suppression of contact hypersensitivity (CHS) after exposure to ultraviolet (UV) radiation and that the antigen-presenting cell (APC) is an important target for this DNA damage.


Subject(s)
DNA Damage , DNA/radiation effects , Dermatitis, Contact/genetics , Immunosuppression Therapy , Ultraviolet Rays , Animals , Cells, Cultured , Dermatitis, Contact/immunology , Humans , Models, Genetic , Models, Immunological
18.
J Immunol ; 161(6): 2684-91, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9743324

ABSTRACT

Considerable evidence suggests that ultraviolet-B (UV-B) radiation suppresses certain immune responses through the induction of cyclobutane pyrimidine dimers in DNA. To determine whether induction of other forms of DNA damage in the skin mimicked the immunosuppressive effects of UV-B radiation, we produced double-strand breaks in the DNA of epidermal cells with HindIII restriction endonuclease encapsulated in liposomes. Application of these liposomes, but not liposomes containing inactive HindIII or an irrelevant endonuclease, to the skin of C3H mice suppressed the induction of delayed-type hypersensitivity responses to Candida albicans and alloantigen and induced IL-10 production in the epidermis. Treatment of the Pam212 murine keratinocyte cell line with these liposomes in vitro induced immunosuppressive activity and IL-10 in culture supernatants. Unlike UV-B irradiation, however, HindIII in liposomes failed to induce suppressor T cell activity in vivo or in vitro. We conclude that double-strand breaks in DNA of epidermal cells can induce immunosuppression and up-regulate the production of immunomodulatory cytokines; however, either DNA damage alone does not account for all the immunosuppressive properties of UV-B irradiation, or cyclobutane pyrimidine dimers differ qualitatively from double-strand breaks in their biologic consequences. These studies raise the possibility that drugs causing DNA damage may induce cytokine dysregulation and immune suppression in addition to cytotoxicity.


Subject(s)
Deoxyribonuclease HindIII/pharmacology , Epidermis/immunology , Hypersensitivity, Delayed/immunology , Immunosuppressive Agents/pharmacology , Interleukin-10/biosynthesis , Liposomes/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Candida albicans/immunology , Cell Line , Cell-Free System/immunology , DNA Damage/immunology , Deoxyribonuclease HindIII/antagonists & inhibitors , Deoxyribonuclease HindIII/immunology , Epidermal Cells , Epidermis/metabolism , Female , Hypersensitivity, Delayed/genetics , Immunosuppressive Agents/antagonists & inhibitors , Interleukin-10/immunology , Isoantigens/immunology , Keratinocytes/cytology , Liposomes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Ultraviolet Rays
19.
J Investig Dermatol Symp Proc ; 3(1): 52-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9732059

ABSTRACT

Ultraviolet (UV) radiation is a potent human carcinogen and it induces skin cancer in experimental animals. Recent studies have shown that unique mutations in the p53 tumor suppressor gene contribute to the development of human and mouse UV-induced skin cancers. Such mutations are also found in sun-damaged skin and actinic keratosis, suggesting that p53 mutations arise early during UV skin carcinogenesis. Our studies have shown that p53 mutations can be detected in UV-irradiated mouse skin months before the gross appearance of skin tumors, suggesting that p53 mutations can serve as a surrogate early biologic endpoint in skin cancer prevention studies. Indeed, application of sun protection factor 15 sunscreens to mouse skin before each UV irradiation resulted in an 88-92% reduction in the number of p53 mutations. Because p53 mutations represent an early essential step in photocarcinogenesis, these results imply that inhibition of this event may protect against skin cancer development.


Subject(s)
Mutation/drug effects , Skin Neoplasms/prevention & control , Skin/radiation effects , Sunscreening Agents/pharmacology , Sunscreening Agents/therapeutic use , Tumor Suppressor Protein p53/genetics , Animals , Humans , Mice , Mutation/radiation effects , Skin Neoplasms/genetics , Ultraviolet Rays
20.
J Immunol ; 161(3): 1327-35, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9686595

ABSTRACT

Cutaneous exposure to low dose (2 kJ/m2) ultraviolet B radiation impairs the induction of contact hypersensitivity (CHS) responses to haptens applied to UV-irradiated skin and induces hapten-specific suppressor T lymphocytes (Ts). Cells collected from the draining lymph nodes of UV-irradiated, FITC-sensitized mice have impaired Ag-presenting activity and induce Ts cells upon injection into syngeneic recipients. This study investigates whether Ts cells originate in the UV-irradiated donor mice or are induced in lymph node cell recipients and the mechanism of suppression. Using congenic mice, we determined that the Ts cells in recipient animals were derived from T cells in the draining lymph nodes of the UV-irradiated donors. Cell lines and clones established from unirradiated and UV-irradiated, FITC-sensitized mice were CD4+, CD8-, TCR-alpha/beta+, MHC restricted, and hapten specific. The T cells proliferated in response to APC sensitized in vivo, but not to APC coupled in vitro with FITC. Cell lines from unirradiated mice were Th1 like, producing large amounts of IFN-gamma, but little IL-4 or IL-10, whereas cloned Ts cells from UV-irradiated mice produced IL-10, but no IL-4 or IFN-gamma. Ts cells blocked APC functions and IL-12 production in vitro. Injection of 5 x 10(4) cloned Ts cells into untreated recipients suppressed the induction of CHS. These results suggest that UV radiation can induce a distinct T regulatory type 1-like Ts population that may block the activation of Th1 cell-mediated immune responses.


Subject(s)
T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/radiation effects , Ultraviolet Rays , Animals , Antigen Presentation/radiation effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/radiation effects , Cell Culture Techniques , Cell Line , Cell Lineage/radiation effects , Female , Immunophenotyping , Lymph Nodes/cytology , Lymphocyte Transfusion , Major Histocompatibility Complex/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Spleen/cytology , T-Lymphocytes, Regulatory/transplantation , Thy-1 Antigens/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...