Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Carcinogenesis ; 33(7): 1360-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22542595

ABSTRACT

Ubiquitous pro-oxidative stressor ultraviolet B radiation (UVB) to human or mouse skin generates platelet-activating factor (PAF) and novel oxidatively modified glycerophosphocholines (Ox-GPCs) with PAF-receptor (PAF-R) agonistic activity. These lipids mediate systemic immunosuppression in a process involving IL-10. The current studies sought to determine the functional significance of UVB-mediated systemic immunosuppression in an established model of murine melanoma. We show that UVB irradiation augments B16F10 tumor growth and is dependent on host, but not melanoma cell; PAF-R-expression as UVB or the PAF-R agonist, carbamoyl PAF (CPAF), both promote B16F10 tumor growth in wild-type (WT) mice, independent of whether B16F10 cells express PAF-Rs, but do not augment tumor growth in Pafr -/- mice. UVB-mediated augmentation of experimental murine tumor growth was inhibited with antioxidants, demonstrating the importance of Ox-GPC PAF-R agonists produced non-enzymatically. Host immune cells are required as CPAF-induced augmentation of tumor growth which is not seen in immunodeficient NOD SCID mice. Finally, depleting antibodies against IL-10 in WT mice or depletion of CD25-positive cells in FoxP3(EGFP) transgenic mice block UVB and/or CPAF-induced tumor growth supporting a requirement for IL-10 and Tregs in this process. These findings indicate that UVB-generated Ox-GPCs with PAF-R agonistic activity enhance experimental murine melanoma tumor growth through targeting host immune cells, most notably Tregs, to mediate systemic immunosuppression.


Subject(s)
Melanoma, Experimental/immunology , Platelet Activating Factor/agonists , Ultraviolet Rays , Animals , Melanoma, Experimental/pathology , Mice , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology
2.
Int J Cancer ; 131(7): E1055-66, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22467332

ABSTRACT

Recent studies suggest that peroxisome proliferator-activated receptor gamma (PPARγ) agonists may have cancer chemopreventive activity. Other studies have shown that loss of epidermal PPARγ results in enhanced chemical carcinogenesis in mice via unknown mechanisms. However, ultraviolet B (UVB) exposure represents the primary etiological agent for skin cancer formation and the role of PPARγ in photobiology and photocarcinogenesis is unknown. In previous studies, we demonstrated that UVB irradiation of cells results in the formation of oxidized glycerophosphocholines that exhibit PPARγ ligand activity. We therefore hypothesized that PPARγ would prove to be a chemopreventive target in photocarcinogenesis. We first showed that UVB irradiation of mouse skin causes generation of PPARγ agonist species in vivo. We then generated SKH-1 hairless, albino mice deficient in epidermal Pparg (Pparg-/-(epi)) using a cytokeratin 14 driven Cre-LoxP strategy. Using a chronic model of UVB photocarcinogenesis, we next showed that Pparg-/-(epi) mice exhibit an earlier onset of tumor formation, increased tumor burden and tumor progression. Increased tumor burden in Pparg-/-(epi) mice was accompanied by a significant increase in epidermal hyperplasia and p53 positive epidermal cells in surrounding skin lacking tumors. After acute UVB irradiation, Pparg-/-(epi) mice exhibited an augmentation of both UVB-induced Caspase 3/7 activity and inflammation. Increased apoptosis and inflammation was also observed after treatment with the PPARγ antagonist GW9662. With chronic UVB irradiation, Pparg-/-(epi) mice exhibited a sustained increase in erythema and transepidermal water loss relative to wildtype littermates. This suggests that PPARγ agonists could have possible chemopreventive activity in non-melanoma skin cancer.


Subject(s)
Apoptosis/radiation effects , Cell Transformation, Neoplastic , Epidermis/metabolism , Epidermis/radiation effects , PPAR gamma/genetics , Skin Neoplasms/metabolism , Ultraviolet Rays/adverse effects , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Epidermis/pathology , Erythema/metabolism , Erythema/pathology , Female , Hyperplasia , Inflammation/metabolism , Inflammation/pathology , Keratinocytes/metabolism , Keratinocytes/pathology , Ligands , Mice , Mice, Hairless , Mice, Knockout , PPAR gamma/deficiency , Skin Neoplasms/pathology , Tumor Burden , Tumor Suppressor Protein p53/metabolism
3.
Carcinogenesis ; 33(3): 694-701, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22223848

ABSTRACT

Although platelet-activating factor (PAF) is a well-known acute inflammatory mediator, little is known regarding the role of PAF in chronic inflammation. Phorbol esters are known to stimulate PAF production. Moreover, the ability of repeated applications of phorbol esters to induce a sustained inflammatory response is crucial to their tumorigenic activity. We therefore examined whether PAF acts as a mediator of phorbol ester-induced inflammation and tumorigenesis. While PAF receptor knockout mice (PAFR(-/-)) showed an expected but modest reduction in the acute inflammatory response to phorbol 12-myristate 13-acetate (PMA), these mice exhibited a surprising increase in inflammation following chronic PMA application. This increased inflammation was documented by a number of findings that included: increased skin thickness, increased myeloperoxidase activity and expression and increased expression of known inflammatory mediators. Interestingly, vehicle-treated PAFR(-/-) mice also exhibited modest increases in levels of inflammatory markers. This suggests that the platelet activating factor receptor (PAFR) acts to suppress chronic inflammation in response to other stimuli, such as barrier disruption. The idea that chronic PAFR activation is anti-inflammatory was documented by repetitive topical PAFR agonist administration that resulted in reduced myeloperoxidase activity in skin. We next utilized a 7,12-dimethylbenz(a)anthracene/PMA carcinogenesis protocol to demonstrate that PAFR(-/-) mice exhibit significantly increased tumor formation and malignant progression compared with wild-type control mice. These studies provide evidence for two important, unexpected and possibly interrelated pathological roles for the PAFR: first, the PAFR acts to suppress PMA-induced chronic inflammation; secondly, the PAFR acts to suppress neoplastic development in response to chemical carcinogens.


Subject(s)
Inflammation/chemically induced , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Skin Neoplasms/chemically induced , Skin/metabolism , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Animals , Cell Transformation, Neoplastic/chemically induced , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Skin/drug effects , Skin/pathology , Tetradecanoylphorbol Acetate/pharmacology
4.
Arch Dermatol Res ; 304(1): 65-71, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21959772

ABSTRACT

Atopic dermatitis (AD) is a pruritic, chronic inflammatory skin disease that affects 10-20% of children and 1-3% of adults worldwide. Recent studies have indicated that the ability of Th2 cytokines, such as interleukin-4 (IL-4) to regulate skin barrier function may be a predisposing factor for AD development. The present studies examined the ability of increased Th2 activity to affect cutaneous barrier function in vivo and epidermal thickening. Mice that express a constitutively active Signal Transducer and Activator of Transcription 6 (STAT6VT) have increased Th2 cells and a predisposition to allergic inflammation were used in these studies, they demonstrate that topical treatment with the irritant sodium lauryl sulfate (SLS) caused increased transepidermal water loss and epidermal thickening in STAT6VT mice over similarly treated wild-type mice. The proliferation marker Ki-67 was increased in the epidermis of STAT6VT compared to the wild-type mice. However, these differences do not appear to be linked to the addition of an irritant as control-treated STAT6VT skin also exhibited elevated Ki-67 levels, suggesting that the increased epidermal thickness in SLS-treated STAT6VT mice is primarily driven by epidermal cell hypertrophy rather than an increase in cellular proliferation. Our results suggest that an environment with increased Th2 cytokines results in abnormal responses to topical irritants.


Subject(s)
Dermatitis, Atopic/immunology , STAT6 Transcription Factor/metabolism , Skin/pathology , T-Lymphocyte Subsets/metabolism , Th2 Cells/metabolism , Animals , Cell Proliferation , Cellular Microenvironment , Dermatitis, Atopic/chemically induced , Genetic Predisposition to Disease , Hypertrophy , Irritants/toxicity , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , Sodium Dodecyl Sulfate/toxicity , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/pathology , Th1-Th2 Balance , Th2 Cells/drug effects , Th2 Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...