Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 343
Filter
1.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34504008

ABSTRACT

Chronic ultraviolet (UV) radiation exposure is the greatest risk factor for cutaneous squamous cell carcinoma (cSCC) development, and compromised immunity accelerates this risk. Having previously identified that epidermal Langerhans cells (LC) facilitate the expansion of UV-induced mutant keratinocytes (KC), we sought to more fully elucidate the immune pathways critical to cutaneous carcinogenesis and to identify potential targets of intervention. Herein, we reveal that chronic UV induces and LC enhance a local immune shift toward RORγt+ interleukin (IL)-22/IL-17A-producing cells that occurs in the presence or absence of T cells while identifying a distinct RORγt+ Sca-1+ CD103+ ICOS+ CD2+/- CCR6+ intracellular CD3+ cutaneous innate lymphoid cell type-3 (ILC3) population (uvILC3) that is associated with UV-induced mutant KC growth. We further show that mutant KC clone size is markedly reduced in the absence of RORγt+ lymphocytes or IL-22, both observed in association with expanding KC clones, and find that topical application of a RORγ/γt inhibitor during chronic UV exposure reduces local expression of IL-22 and IL-17A while markedly limiting mutant p53 KC clonal expansion. We implicate upstream Toll-like receptor signaling in driving this immune response to chronic UV exposure, as MyD88/Trif double-deficient mice also show substantially reduced p53 island number and size. These data elucidate key immune components of chronic UV-induced cutaneous carcinogenesis that might represent targets for skin cancer prevention.


Subject(s)
Interleukins/metabolism , Keratinocytes/pathology , Lymphocytes/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Skin Neoplasms/pathology , Skin/pathology , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Cells, Cultured , Immunity, Innate/immunology , Interleukins/genetics , Keratinocytes/metabolism , Keratinocytes/radiation effects , Langerhans Cells/immunology , Langerhans Cells/metabolism , Langerhans Cells/pathology , Langerhans Cells/radiation effects , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/radiation effects , Mice , Mutation , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Skin/metabolism , Skin/radiation effects , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Interleukin-22
2.
Toxicol In Vitro ; 53: 193-199, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30149078

ABSTRACT

Lifestyle changes involving frequent outdoor activities are contributing to higher exposure to harmful ultraviolet light (UVB). The acute effects of UVB irradiation on human skin was evaluated in this study using freshly excised human skin from elective surgery subjected to UVB doses (0-3.76 J/cm2). The assessment of UVB induced cellular and skin damages was undertaken at two time points immediately and 24 h post exposure using in vitro, and immunohistochemical staining techniques. The results indicated no significant loss of skin integrity or significant acute mitochondrial cellular damages in UVB exposed skin sections as measured by the MTS cytotoxicity assay. The other key markers of damage showed significant extracellular LDH membrane leakages and upregulation of inflammatory cytokines such as IL-1ß. Skin integrity analysis was also undertaken using H&E, HLADR, and anti-cytokeratin antibodies. The results showed significant epidermal changes, basal cell activation and Langerhans cells depletion. The research proved the usefulness of freshly excised human skin explant model in measuring UVB damage. Furthermore, freshly excised human skin maintains the natural layering and therefore does not pose the same challenges faced by commercially available reconstructed skin in terms of higher costs and accurate mimicking of all the complex interactions observed in human skin.


Subject(s)
Skin/radiation effects , Ultraviolet Rays/adverse effects , Cell Line , Humans , Interleukin-1beta/metabolism , L-Lactate Dehydrogenase/metabolism , Langerhans Cells/radiation effects , Skin/metabolism , Skin/pathology
3.
Sci Transl Med ; 10(454)2018 08 15.
Article in English | MEDLINE | ID: mdl-30111646

ABSTRACT

Photosensitivity, or skin sensitivity to ultraviolet radiation (UVR), is a feature of lupus erythematosus and other autoimmune and dermatologic conditions, but the mechanistic underpinnings are poorly understood. We identify a Langerhans cell (LC)-keratinocyte axis that limits UVR-induced keratinocyte apoptosis and skin injury via keratinocyte epidermal growth factor receptor (EGFR) stimulation. We show that the absence of LCs in Langerin-diphtheria toxin subunit A (DTA) mice leads to photosensitivity and that, in vitro, mouse and human LCs can directly protect keratinocytes from UVR-induced apoptosis. LCs express EGFR ligands and a disintegrin and metalloprotease 17 (ADAM17), the metalloprotease that activates EGFR ligands. Deletion of ADAM17 from LCs leads to photosensitivity, and UVR induces LC ADAM17 activation and generation of soluble active EGFR ligands, suggesting that LCs protect by providing activated EGFR ligands to keratinocytes. Photosensitive systemic lupus erythematosus (SLE) models and human SLE skin show reduced epidermal EGFR phosphorylation and LC defects, and a topical EGFR ligand reduces photosensitivity. Together, our data establish a direct tissue-protective function for LCs, reveal a mechanistic basis for photosensitivity, and suggest EGFR stimulation as a treatment for photosensitivity in lupus erythematosus and potentially other autoimmune and dermatologic conditions.


Subject(s)
Cytoprotection/radiation effects , Keratinocytes/cytology , Keratinocytes/radiation effects , Langerhans Cells/cytology , Langerhans Cells/radiation effects , Ultraviolet Rays , ADAM17 Protein/metabolism , Animals , Apoptosis/radiation effects , Disease Models, Animal , Epidermis/metabolism , Epidermis/radiation effects , ErbB Receptors/metabolism , Humans , Ligands , Lupus Erythematosus, Systemic/pathology , Mice, Inbred C57BL , Phosphorylation/radiation effects
5.
Photochem Photobiol ; 94(1): 150-157, 2018 01.
Article in English | MEDLINE | ID: mdl-28763105

ABSTRACT

Phototherapy is routinely used for the treatment of various skin conditions and targeted therapy of superficial cancers. However, the molecular mechanisms behind their biological effects and the need for efficacy enhancing photosensitizers are not well addressed. Particularly, not much is known about the inherent effect of light from the visible spectrum on cytokine release and its downstream effects in keratinocytes and immune cells located in skin and therefore exposed to light. To address this, we delivered calibrated doses of well-defined light qualities (380 to 660 nm) to cocultures of human keratinocytes and macrophage/dendritic cells in the absence or presence of the commonly used photosensitizer 8-methoxypsoralen (8-MOP). The experiments identified IL-4 as a key effector cytokine released by this coculture model with need for 8-MOP in the UVA1 /blue (380 nm) and no requirement for photosensitizer in the red light spectrum (627 nm). 3D organotypic skin cultures treated with IL-4 showed thickening of the epidermal layer and delayed differentiation. However unlike IL-4 and UVA1 /blue light treatment, red light did not reduce the expression of keratinocyte differentiation markers or increase signs of photo-oxidative damage. This supports the application of isolated red light as a possible alternative for photo-immunotherapy without need for additional photosensitizers.


Subject(s)
Interleukin-4/metabolism , Keratinocytes/immunology , Keratinocytes/radiation effects , Langerhans Cells/immunology , Langerhans Cells/radiation effects , Cell Differentiation/immunology , Cell Line , Coculture Techniques , Humans , Keratinocytes/drug effects , Langerhans Cells/drug effects , Light , Methoxsalen/pharmacology , Photosensitizing Agents/pharmacology , Phototherapy/methods , Reactive Oxygen Species/metabolism , THP-1 Cells
6.
Arterioscler Thromb Vasc Biol ; 37(1): 66-74, 2017 01.
Article in English | MEDLINE | ID: mdl-27765767

ABSTRACT

OBJECTIVE: UVB irradiation is an established treatment for immunoinflammatory cutaneous disorders and has been shown to suppress cutaneous and systemic inflammatory diseases through modulation of the adaptive immune response. However, it remains unknown whether UVB irradiation prevents an immunoinflammatory disease of arteries such as atherosclerosis. APPROACH AND RESULTS: Here, we show that UVB exposure inhibits the development and progression of atherosclerosis in atherosclerosis-prone mice by expanding and enhancing the functional capacity of CD4+ forkhead box P3+ regulatory T cells and regulating proatherogenic T-cell responses. Experimental studies in Langerhans cell-depleted mice revealed that epidermal Langerhans cells play a critical role in UVB-dependent induction of CD4+ forkhead box P3+ regulatory T cells, suppression of proatherogenic T-cell responses, and prevention of atherosclerotic plaque development. CONCLUSIONS: Our findings suggest the skin immune system as a novel therapeutic target for atherosclerosis and provide a novel strategy for the treatment and prevention of atherosclerosis.


Subject(s)
Aorta/radiation effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Inflammation/prevention & control , Skin/radiation effects , T-Lymphocytes, Regulatory/radiation effects , Ultraviolet Rays , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Genetic Predisposition to Disease , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Langerhans Cells/immunology , Langerhans Cells/metabolism , Langerhans Cells/radiation effects , Lymphocyte Activation/radiation effects , Mice, Knockout , Phenotype , Plaque, Atherosclerotic , Signal Transduction/radiation effects , Skin/immunology , Skin/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
10.
J Photochem Photobiol B ; 155: 98-103, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26774380

ABSTRACT

Infrared irradiation (IR) is the most abundant fraction of sunlight reaching the earth's surface and provides heat. The fever response of an animal is known to regulate its immune responses. However, the non-thermal immune responses of IR were difficult to assess owing to its close association with heat. We hypothesized that IR irradiation induced differential immunological responses, independent of its associated heat. With an IR machine coupled with a delicate temperature control system, we investigated the non-thermal immunological effects of IR in vivo. With heating at 37 °C or 39 °C using an electric blanket or IR irradiation, we measured the skin's physiological parameters, including transepidermal water loss (TEWL), pH, skin hydration, elasticity, sebum production, and skin blood flow. We also measured the number of Langerhans cells in epidermal sheets and draining lymph nodes. Lymph node cells were activated by anti-CD3 antibody and their production of interleukin (IL)-5, 10, 13, 17, and interferon (IFN)-γ was measured by enzyme-linked immunosorbent assay (ELISA). The result showed that compared to heating alone, IR causes an enhanced activation of epidermal Langerhans cells, both in epidermal sheets and in draining lymph nodes. The activation of draining lymph node cells by anti-CD3 antibody in vitro induces both Th2 and Th1, but not Treg immune responses. Interestingly, IL-13, a Th2 cytokine, is induced the most. In contrast, physiological parameters and barrier functions of skin were not altered after IR irradiation. The study showed that IR alone without heat modulates immune responses in vivo, indicating that IR irradiation might regulate host immunity in a heat-independent manner.


Subject(s)
Infrared Rays , Skin/radiation effects , Animals , Antibodies/immunology , Antibodies/pharmacology , CD3 Complex/immunology , Cytokines/analysis , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Langerhans Cells/immunology , Langerhans Cells/radiation effects , Lymph Nodes/cytology , Lymph Nodes/metabolism , Lymph Nodes/radiation effects , Mice , Mice, Inbred C57BL , Skin/chemistry , Skin/immunology , Skin Temperature/radiation effects , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Water/chemistry
12.
Nat Immunol ; 16(10): 1060-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26343536

ABSTRACT

Treatment with ionizing radiation (IR) can lead to the accumulation of tumor-infiltrating regulatory T cells (Treg cells) and subsequent resistance of tumors to radiotherapy. Here we focused on the contribution of the epidermal mononuclear phagocytes Langerhans cells (LCs) to this phenomenon because of their ability to resist depletion by high-dose IR. We found that LCs resisted apoptosis and rapidly repaired DNA damage after exposure to IR. In particular, we found that the cyclin-dependent kinase inhibitor CDKN1A (p21) was overexpressed in LCs and that Cdkn1a(-/-) LCs underwent apoptosis and accumulated DNA damage following IR treatment. Wild-type LCs upregulated major histocompatibility complex class II molecules, migrated to the draining lymph nodes and induced an increase in Treg cell numbers upon exposure to IR, but Cdkn1a(-/-) LCs did not. Our findings suggest a means for manipulating the resistance of LCs to IR to enhance the response of cutaneous tumors to radiotherapy.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Langerhans Cells/radiation effects , Radiation, Ionizing , T-Lymphocytes, Regulatory/radiation effects , Animals , Cell Survival/genetics , Cell Survival/radiation effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Flow Cytometry , Mice , Microarray Analysis , Polymerase Chain Reaction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Up-Regulation
13.
J Invest Dermatol ; 135(11): 2824-2833, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26053049

ABSTRACT

UVB light is considered the major environmental inducer of human keratinocyte (KC) DNA mutations, including within the tumor-suppressor gene p53, and chronic exposure is associated with cutaneous squamous cell carcinoma formation. Langerhans cells (LCs) comprise a dendritic network within the suprabasilar epidermis, yet the role of LCs in UVB-induced carcinogenesis is largely unknown. Herein we show that LC-intact epidermis develops UVB-induced tumors more readily than LC-deficient epidermis. Although levels of epidermal cyclopyrimidine dimers following acute UVB exposure are equivalent in the presence or absence of LCs, chronic UVB-induced p53 mutant clonal islands expand more readily in association with LCs, which remain largely intact and are preferentially found in proximity to the expanding mutant KC populations. The observed LC facilitation of mutant p53 clonal expansion is completely αß and γδ T-cell independent and is associated with increased intraepidermal expression of IL-22 and the presence of group 3 innate lymphoid cells. These data demonstrate that LCs have a key role in UVB-induced cutaneous carcinogenesis and suggest that LCs locally stimulate KC proliferation and innate immune cells that provoke tumor outgrowth.


Subject(s)
Carcinogenesis/pathology , Cell Proliferation/radiation effects , Epidermis/radiation effects , Langerhans Cells/radiation effects , Skin Neoplasms/etiology , Ultraviolet Rays/adverse effects , Animals , Biopsy, Needle , Cells, Cultured , Disease Models, Animal , Epidermis/pathology , Female , Flow Cytometry , Gene Expression Regulation , Humans , Immunohistochemistry , Interleukins/metabolism , Interleukins/radiation effects , Langerhans Cells/pathology , Mice , Mice, Inbred Strains , Skin Neoplasms/pathology , Interleukin-22
14.
Drug Des Devel Ther ; 8: 1817-25, 2014.
Article in English | MEDLINE | ID: mdl-25336924

ABSTRACT

BACKGROUND: Topical pimecrolimus has been shown to reverse epidermal CD1a(+) Langerhans cell reduction induced by high-dose ultraviolet (UV)B irradiation, but the mechanism is still unclear. This study aimed to investigate the possible mechanism of the effect of pimecrolimus on high-dose UVB-irradiated epidermal Langerhans cells. METHODS: FORTY HUMAN FORESKIN TISSUES WERE DIVIDED INTO FOUR GROUPS: control; pimecrolimus-only; UVB-only; and UVB + pimecrolimus. All tissues were cultured, and each tissue was cut into four pieces, corresponding to four time points (0 hours, 18 hours, 24 hours, and 48 hours). We collected the tissues and culture medium at each time point. The percentage of CD1a(+) cells in medium was detected by flow cytometry. The tissues were detected for messenger (m)RNA and protein expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and E-cadherin, by reverse-transcription polymerase chain reaction (PCR) and Western blot. RESULTS: At 18 hours, 24 hours, and 48 hours, the CD1a(+) cells in the culture medium of the UVB-only group and the UVB + pimecrolimus group were significantly more than in the control group, while the CD1a(+) cells of the UVB + pimecrolimus group was less than of the UVB-only group. For both the UVB-only group and UVB + pimecrolimus group, TNF-α expression (by both reverse-transcription PCR and Western blot) of the tissues was clearly higher and E-cadherin expression was significantly lower compared with the control group, at 18 hours, 24 hours, and 48 hours. For the UVB + pimecrolimus group, TNF-α was clearly lower and E-cadherin was significantly higher compared with the UVB-only group. CONCLUSION: Topical pimecrolimus inhibited epidermal Langerhans cell migration induced by high-dose UVB irradiation, via regulation of TNF-α and E-cadherin.


Subject(s)
Cadherins/metabolism , Cell Movement/drug effects , Cell Movement/radiation effects , Langerhans Cells/drug effects , Tacrolimus/analogs & derivatives , Tumor Necrosis Factor-alpha/metabolism , Ultraviolet Rays , Administration, Topical , Adolescent , Adult , Calcineurin Inhibitors/administration & dosage , Calcineurin Inhibitors/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Langerhans Cells/cytology , Langerhans Cells/metabolism , Langerhans Cells/radiation effects , Structure-Activity Relationship , Tacrolimus/administration & dosage , Tacrolimus/pharmacology , Young Adult
15.
Dermatol Surg ; 40(9): 979-87, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25072126

ABSTRACT

BACKGROUND: Photodynamic therapy (PDT) using aminolevulinic acid (ALA) with blue light or red light is effective for treating actinic keratoses (AKs). However, immunosuppression follows red light PDT, raising the spectre of skin cancer promotion in treated skin. OBJECTIVE: To determine whether broad-area short incubation (BASI)-ALA-PDT using blue light immunosuppression immunosuppresses treated skin. METHODS: Patients were evaluated clinically and by standardized facial biopsies of non-AK skin before, 24 hours and 1 month after customary blue light BASI-ALA-PDT. All biopsies were stained for markers of epidermal atypia and Langerhans cells (LCs); and at 24 hours to detect oxidative DNA damage. RESULTS: Patients had an 81% reduction in AKs and slight improvement in clinical and histologic signs of photoaging after 1 month. The biopsied chronically photodamaged skin without clinically detectable AKs showed no effect of PDT on the LC number, distribution, or morphology; and no oxidative DNA damage, in contrast to the changes reported after customary red light PDT. CONCLUSION: Customary blue light BASI-ALA-PDT does not affect the LC number or produce oxidative DNA damage, the sequelae of red light PDT responsible for immunosuppression in treated skin.


Subject(s)
DNA Damage , Keratosis, Actinic/drug therapy , Langerhans Cells/radiation effects , Photochemotherapy/methods , Skin Aging/radiation effects , Aged , Aminolevulinic Acid/pharmacology , Cell Count , Color , Female , Humans , Immunosuppression Therapy , Keratinocytes/chemistry , Keratosis, Actinic/pathology , Ki-67 Antigen/analysis , Langerhans Cells/drug effects , Male , Middle Aged , Oxidative Stress , Photochemotherapy/adverse effects , Photosensitizing Agents/pharmacology , Skin Aging/drug effects , Skin Aging/pathology , Tumor Suppressor Protein p53/analysis
16.
Exp Dermatol ; 23(6): 428-30, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24758562

ABSTRACT

The pathogenesis of polymorphic light eruption (PLE) has been linked to a lack of UV-induced immune suppression. To determine the role of Langerhans cells (LC), mast cells and regulatory T cells, biopsies from PLE patients were taken from exposed sites in spring before and after photohardening with 311 nm or PUVA as well as again in summer. Skin sections were assessed for the presence of Langerin/CD1a+ LC and CD3+, CD4+, CD25+ or FoxP3+ T cells and mast cells. Photohardening transiently decreased the density of epidermal LC and significantly increased a low baseline mast cell density in the papillary dermis of PLE patients. Baseline T cell numbers in the skin were low, and there was no difference in PLE patients among any time point. This suggests that LC suppression together with recruitment of mast cells into photohardened skin may be a key cellular event underlying the mechanism by which phototherapy protects from PLE.


Subject(s)
Dermis/pathology , Langerhans Cells/pathology , Mast Cells/pathology , Photosensitivity Disorders/pathology , Photosensitivity Disorders/therapy , Phototherapy , Skin Diseases, Genetic/pathology , Skin Diseases, Genetic/therapy , Ultraviolet Rays , Adult , Biopsy , Case-Control Studies , Cell Count , Dermis/radiation effects , Female , Humans , Langerhans Cells/radiation effects , Mast Cells/radiation effects , Middle Aged , PUVA Therapy , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/radiation effects , Treatment Outcome
17.
Kobe J Med Sci ; 59(1): E1-9, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23756657

ABSTRACT

Narrowband ultraviolet B (NB-UVB) induces different immunological features from broadband ultraviolet B and is effective for the treatment of various cutaneous diseases. UV exposure alters the morphology and function of epidermal Langerhans cells (LCs), which can elicit cutaneous immunosuppressive responses. Recent studies have proposed that LCs serve as immunoregulatory cells in UV-induced immune suppression. This study investigated the cellular mechanisms of NB-UVB-induced immune suppression, including its effects on LC migration. NB-UVB irradiation induced the migration of epidermal LCs from the skin to the draining lymph nodes in a time- and dose-dependent manner. Experiments in Lang-DTR knock-in mice confirmed that epidermal LCs rather than Langerin+ dermal dendritic cells are essential for NB-UVB-induced immune suppression. These findings indicate that LCs play a critical immunoregulatory role in NB-UVB-induced immune suppression and NB-UVB phototherapy.


Subject(s)
Immune Tolerance/radiation effects , Langerhans Cells/radiation effects , Ultraviolet Rays , Animals , Cell Movement/radiation effects , Dose-Response Relationship, Radiation , Female , Langerhans Cells/physiology , Mice , Mice, Inbred C57BL , Ultraviolet Therapy
18.
Photochem Photobiol ; 89(5): 1249-54, 2013.
Article in English | MEDLINE | ID: mdl-23663071

ABSTRACT

To examine the effect of laser thermal injury on Langerhans cells (LC) within the epidermis, the dorsal skin of mice and hairless guinea pigs was exposed to varying levels of laser irradiation using a thulium laser at a wavelength of 2.0 µm. At 6, 24 and 48 h post irradiation, animals were euthanized, skin samples prepared for histology and the epidermis obtained and stained by major histocompatibility complex-II staining (mice) or ATPase assay (hairless guinea pigs) for the enumeration of LC. Mouse skin exhibited histological evidence of thermal damage at 24 h post irradiation at even the lowest dose (0.14 W) and decreases in the numbers of epidermal LC were observed at all doses and decreases were proportional to dose. In contrast, hairless guinea pig skin only showed consistent histological evidence of thermal damage at the highest dose of irradiation (0.70 W) at 24 and 48 h post irradiation and exhibited a statistically significant decrease in numbers of epidermal LC only at this dose. Thus, epidermal LC depletion occurred in the skin of both mice and hairless guinea pigs in response to laser treatment and the magnitude of depletion directly correlated with the extent of thermal damage both within and between species.


Subject(s)
Epidermis/radiation effects , Langerhans Cells/radiation effects , Lasers , Adenosine Triphosphatases/metabolism , Animals , Epidermal Cells , Female , Guinea Pigs , Histocompatibility Antigens Class II/metabolism , Mice , Mice, Inbred BALB C
20.
Nanotoxicology ; 7(8): 1386-98, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23078247

ABSTRACT

Ultraviolet radiation (UVR) skin exposure is a common exogenous insult that can alter skin barrier and immune functions. With the growing presence of nanoparticles (NPs) in consumer goods and technological applications the potential for NPs to contact UVR-exposed skin is increasing. Therefore it is important to understand the effect of UVR on NP skin penetration and the potential for systemic translocation. Previous studies qualitatively showed that UVR skin exposure can increase the penetration of NPs below the stratum corneum. In this work, an in vivo mouse model was used to quantitatively examine the skin penetration of carboxylated (CdSe/ZnS, core/shell) quantum dots (QDs) through intact and UVR barrier-disrupted murine skin by organ Cd mass analysis. Transepidermal water loss was used to measure the magnitude of the skin barrier defect as a function of UVR dose and time post-UVR exposure. QDs were applied to mice 3-4 days post-UVR exposure at the peak of the skin barrier disruption. Our results reveal unexpected trends that suggest these negative-charged QDs can penetrate barrier intact skin and that penetration and systemic transport depends on the QD application time post-UVR exposure. The effect of UVR on skin-resident dendritic cells and their role in the systemic translocation of these QDs are described. Our results suggest that NP skin penetration and translocation may depend on the specific barrier insult and the inflammatory status of the skin.


Subject(s)
Quantum Dots , Skin Absorption/radiation effects , Skin/metabolism , Skin/radiation effects , Ultraviolet Rays , Analysis of Variance , Animals , Female , Langerhans Cells/radiation effects , Male , Mice , Skin/chemistry , Tissue Distribution , Water/analysis , Water/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...