Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Photochem Photobiol Sci ; 14(4): 801-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25645888

ABSTRACT

Ultraviolet (UV) wavelengths in sunlight are the prime cause of skin cancer in humans with both the UVA and UVB wavebands making a contribution to photocarcinogenesis. UV has many different biological effects on the skin that contribute to carcinogenesis, including suppression of adaptive immunity, sunburn and altering the migration of mast cells into and away from irradiated skin. Many molecular mechanisms have been identified as contributing to skin responses to UV. Recently, using gene set enrichment analysis of microarray data, we identified the alternative complement pathway with a central role for factor B (fB) in UVA-induced immunosuppression. In the current study we used mice genetically deficient in fB (fB-/- mice) to study the functional role of the alternative complement pathway in skin responses to UV. We found that fB is required for not only UVA but also UVB-induced immunosuppression and solar-simulated UV induction of the oedemal component of sunburn. Factor B-/- mice had a larger number of resident skin mast cells than control mice, but unlike the controls did not respond to UV by increasing mast cell infiltration into the skin. This study provides evidence for a function role for fB in skin responses to UV radiation. Factor B regulates UVA and UVB induced immunosuppression, UV induced oedema and mast cell infiltration into the skin. The alternative complement pathway is therefore an important regulator of skin responses to UV.


Subject(s)
Complement Factor B/metabolism , Edema/physiopathology , Hypersensitivity, Delayed/physiopathology , Mast Cells/radiation effects , Skin/radiation effects , Ultraviolet Rays/adverse effects , Animals , Cell Movement/physiology , Cell Movement/radiation effects , Complement Factor B/genetics , Disease Models, Animal , Edema/etiology , Female , Hypersensitivity, Delayed/etiology , Male , Mast Cells/physiology , Mice, Inbred C57BL , Mice, Knockout , Skin/physiopathology , Sunburn/etiology , Sunburn/physiopathology
2.
J Neuroinflammation ; 9: 246, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23111065

ABSTRACT

Infiltration of Ly6C(hi) monocytes from the blood is a hallmark of viral encephalitis. In mice with lethal encephalitis caused by West Nile virus (WNV), an emerging neurotropic flavivirus, inhibition of Ly6C(hi) monocyte trafficking into the brain by anti-very late antigen (VLA)-4 integrin antibody blockade at the time of first weight loss and leukocyte influx resulted in long-term survival of up to 60% of infected mice, with subsequent sterilizing immunity. This treatment had no effect on viral titers but appeared to be due to inhibition of Ly6C(hi) macrophage immigration. Although macrophages isolated from the infected brain induced WNV-specific CD4(+) T-cell proliferation, T cells did not directly contribute to pathology, but are likely to be important in viral control, as antibody-mediated T-cell depletion could not reproduce the therapeutic benefit of anti-VLA-4. Instead, 70% of infiltrating inflammatory monocyte-derived macrophages were found to be making nitric oxide (NO). Furthermore, aminoguanidine-mediated inhibition of induced NO synthase activity in infiltrating macrophages significantly prolonged survival, indicating involvement of NO in the immunopathology. These data show for the first time the therapeutic effects of temporally targeting pathogenic NO-producing macrophages during neurotropic viral encephalitis.


Subject(s)
Integrin alpha4beta1/immunology , Integrin alpha4beta1/metabolism , Macrophages/immunology , Macrophages/metabolism , West Nile Fever , Animals , Antigens, CD/metabolism , Brain/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Viral/physiology , Glial Fibrillary Acidic Protein/metabolism , Integrins/genetics , Integrins/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophages/virology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/immunology , Nitric Oxide Synthase Type II , West Nile Fever/immunology , West Nile Fever/metabolism , West Nile Fever/pathology
3.
Glob J Health Sci ; 4(6): 109-18, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-23121747

ABSTRACT

Nepal and Alberta are literally a world apart. Yet they share a common problem of restricted access to health services in remote and rural areas. In Nepal, urban-rural disparities were one of the main issues in the recent civil war, which ended in 2006. In response to the need for improved health equity in Nepal a dedicated group of Nepali physicians began planning the Patan Academy of Health Sciences (PAHS), a new health sciences university dedicated to the education of rural health providers in the early 2000s. Beginning with a medical school the Patan Academy of Health Sciences uses international help to plan, deliver and assess its curriculum. PAHS developed an International Advisory Board (IAB) attracting international help using a model of broad, intentional recruitment and then on individuals' natural attraction to a clear mission of peace-making through health equity. Such a model provides for flexible recruitment of globally diverse experts, though it risks a lack of coordination. Until recently, the PAHS IAB has not enjoyed significant or formal support from any single international institution. However, an increasing number of the international consultants recruited by PAHS to its International Advisory Board are from the University of Alberta in Edmonton, Alberta, Canada (UAlberta). The number of UAlberta Faculty of Medicine and Dentistry members involved in the project has risen to fifteen, providing a critical mass for a coordinated effort to leverage institutional support for this partnership. This paper describes the organic growth of the UAlberta group supporting PAHS, and the ways in which it supports a sister institution in a developing nation.


Subject(s)
International Cooperation , Rural Health Services/organization & administration , Schools, Medical/organization & administration , Canada , Capital Financing , Curriculum , Developing Countries , Humans , Nepal , Socioeconomic Factors , Staff Development , Vital Statistics
4.
Exp Dermatol ; 21(8): 599-604, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22775994

ABSTRACT

Brm is an ATPase subunit of the SWI/SNF chromatin-remodelling complex. Previously, we identified a novel hotspot mutation in Brm in human skin cancer, which is caused by exposure to ultraviolet radiation (UVR). As SWI/SNF is involved in DNA repair, we investigated whether Brm-/- mice had enhanced photocarcinogenesis. P53+/- and Brm-/-p53+/- mice were also examined as the p53 tumor suppressor gene is mutated early during human skin carcinogenesis. Mice were exposed to a low-dose irradiation protocol that caused few skin tumors in wild-type mice. Brm-/- mice with both p53 alleles intact had an increased incidence of skin and ocular tumors compared to Brm+/+p53+/+ controls. Brm loss in p53+/- mice did not further enhance skin or ocular cancer incidence beyond the increased photocarcinogenesis in p53+/- mice. However, the skin tumors that arose early in Brm-/- p53+/- mice had a higher growth rate. Brm-/- did not prevent UVR-induced apoptotic sunburn cell formation, which is a protective response. Unexpectedly, Brm-/- inhibited UVR-induced immunosuppression, which would be predicted to reduce rather than enhance photocarcinogenesis. In conclusion, the absence of Brm increased skin and ocular photocarcinogenesis. Even when one allele of p53 is lost, Brm has additional tumor suppressing capability.


Subject(s)
Eye Neoplasms/physiopathology , Neoplasms, Radiation-Induced/physiopathology , Skin Neoplasms/physiopathology , Transcription Factors/deficiency , Ultraviolet Rays/adverse effects , Animals , Apoptosis , Dose-Response Relationship, Radiation , Eye Neoplasms/genetics , Eye Neoplasms/pathology , Female , Genetic Predisposition to Disease/genetics , Incidence , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/physiology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
5.
J Dermatol Sci ; 66(3): 176-82, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22277701

ABSTRACT

Low doses of sunlight that can be received during normal daily activities suppress immunity in humans. Both ultraviolet (UV) B (290-320 nm) and UVA (320-400 nm) are immunosuppressive. The wavelength dependence in humans shows distinct non-overlapping immunosuppressive peaks of solar effectiveness centred at 310 nm UVB and 370 nm UVA. In murine models of systemic immunosuppression low dose UV inhibits expansion of effector T cells in skin-draining lymph nodes, and retention of dermal effector memory CD8T cells at sites of antigen challenge. In addition to suppressing skin immunity, UV inhibits immunity in internal organs, including activation of CD8 T cells and cytotoxic T cell activity in the spleen, and memory T cell activation in the spleen and bone marrow. Neither of the chromophores responsible for UV suppression of skin immunity, DNA damage and urocanic acid, nor reactive oxygen species are involved in regulation of CD8 T cells in internal organs. Thus UVB impedes the activation and cytotoxicity of antigen-specific T cells in internal organs by mechanisms independent of suppression of skin immunity. These deleterious effects of low dose UV on skin immunity are likely to contribute to skin cancer, however UV suppression of immunity in internal organs may protect from autoimmunity. Epidemiological evidence suggests that sunlight protects from some autoimmune diseases directed towards internal organs. As UV suppression of skin and internal organ immunity appear to occur via different mechanisms, it may be possible to protect skin immunity and therefore reduce skin cancer incidence without preventing UV from reducing autoimmunity in internal organs.


Subject(s)
Autoimmunity/physiology , Skin/immunology , Spleen/immunology , Ultraviolet Rays/adverse effects , Animals , Humans , Immune Tolerance/physiology , Lymphocyte Activation/physiology , Models, Animal , Skin Neoplasms/physiopathology
6.
Am J Pathol ; 178(6): 2783-91, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21641400

ABSTRACT

Exposure to UVB radiation before antigen delivery at an unirradiated site inhibits functional immunological responses. Mice treated dorsally with suberythemal low-dose UVB and immunized with ova in abdominal skin generated ova-specific CD8 T cells with a significantly decreased activation, expansion, and cytotoxic activity compared with unirradiated mice. UVB also impaired the delayed-type hypersensitivity (DTH) reaction to ova. Transfer of CD4⁺CD25⁺cells from UVB-exposed mice did not suppress the ova-specific CD8 T-cell response or DTH reaction in unexposed mice, confirming that systemic low-dose UVB does not induce long-lived functional regulatory CD4⁺CD25⁺ T cells. Repairing cyclobutane pyrimidine dimer-type DNA damage and blocking aryl hydrocarbon receptor signaling also did not reverse the immunosuppressive effect of UVB on ova-specific CD8 T cells and DTH, suggesting that cyclobutane pyrimidine dimers and the aryl hydrocarbon receptor are not required in systemic low-dose UVB-induced immunosuppression. The known UVB chromophore, cis-urocanic acid, and reactive oxygen species triggered the inhibition of DTH caused by UVB, but they were not involved in the modulation of CD8 T cells. These findings indicate that systemic low-dose UVB impedes the primary response of antigen-specific CD8 T cells by a novel mechanism that is independent of pathways known to be involved in systemic suppression of DTH.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Inflammation/pathology , Skin/immunology , Skin/pathology , Ultraviolet Rays , Administration, Topical , Animals , Antioxidants/pharmacology , CD4 Antigens/metabolism , CD8-Positive T-Lymphocytes/drug effects , DNA Repair/drug effects , DNA Repair/radiation effects , Dose-Response Relationship, Radiation , Female , Hypersensitivity, Delayed/complications , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Inflammation/complications , Inflammation/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Pyrimidine Dimers/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Skin/drug effects , Skin/radiation effects , Spleen/drug effects , Spleen/pathology , Spleen/radiation effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects , Urocanic Acid/administration & dosage , Urocanic Acid/pharmacology
7.
Photochem Photobiol Sci ; 9(1): 25-30, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20062841

ABSTRACT

Ultraviolet A (UVA) radiation can have dual affects on the immune system depending on dose. At doses of approximately 1.8 J cm(-2), UVA acts in an immunosuppressive manner, whilst at higher doses UVA can promote recovery and protection against UVB-induced immunosuppression in mice. We utilised a model of contact hypersensitivity (CHS) to investigate how different doses of UVA modulates CD8 T cell immunity against a hapten in vivo. Only 1.8 J cm(-2) UVA decreased the CHS response compared to unirradiated mice, but this did not correlate with an inhibition of primary effector CD8 T cells. A similar expansion of effector CD8 T cells in skin-draining lymph nodes and accumulation of IFN-gamma-producing CD8 T cells in the ear skin was observed between unirradiated and UVA-irradiated mice. However, dermal memory CD8 T cells examined 9 weeks post challenge showed decreased numbers in mice irradiated with 1.8 J cm(-2) UVA compared with unirradiated, 1.3 J cm(-2) and 3.4 J cm(-2) UVA-irradiated mice. Therefore, UVA does not inhibit the expansion, migration or IFN-gamma secretion of CD8 T cells during a primary immune response. However, exposure to immunosuppressive UVA causes a defect in CD8 T cell development that impairs the ability of cells to become long-term memory cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Immune Tolerance/radiation effects , Skin/immunology , Skin/radiation effects , Ultraviolet Rays , Animals , CD8-Positive T-Lymphocytes/cytology , Dermatitis, Contact/immunology , Dose-Response Relationship, Radiation , Female , Mice , Skin/cytology , Time Factors
8.
J Exp Med ; 205(10): 2319-37, 2008 Sep 29.
Article in English | MEDLINE | ID: mdl-18779347

ABSTRACT

In a lethal West Nile virus (WNV) model, central nervous system infection triggered a threefold increase in CD45(int)/CD11b(+)/CD11c(-) microglia at days 6-7 postinfection (p.i.). Few microglia were proliferating, suggesting that the increased numbers were derived from a migratory precursor cell. Depletion of "circulating" (Gr1(-)(Ly6C(lo))CX3CR1(+)) and "inflammatory" (Gr1(hi)/Ly6C(hi)/CCR2(+)) classical monocytes during infection abrogated the increase in microglia. C57BL/6 chimeras reconstituted with cFMS-enhanced green fluorescent protein (EGFP) bone marrow (BM) showed large numbers of peripherally derived (GFP(+)) microglia expressing GR1(+)(Ly6C(+)) at day 7 p.i., suggesting that the inflammatory monocyte is a microglial precursor. This was confirmed by adoptive transfer of labeled BM (Ly6C(hi)/CD115(+)) or circulating inflammatory monocytes that trafficked to the WNV-infected brain and expressed a microglial phenotype. CCL2 is a chemokine that is highly expressed during WNV infection and important in inflammatory monocyte trafficking. Neutralization of CCL2 not only reduced the number of GFP(+) microglia in the brain during WNV infection but prolonged the life of infected animals. Therefore, CCL2-dependent inflammatory monocyte migration is critical for increases in microglia during WNV infection and may also play a pathogenic role during WNV encephalitis.


Subject(s)
Antigens, Ly/immunology , Brain/immunology , Brain/virology , Microglia/immunology , Monocytes/immunology , Stem Cells/immunology , West Nile Fever/immunology , Adoptive Transfer , Animals , Blood-Brain Barrier/physiology , Brain/cytology , Brain/pathology , Chemokine CCL2/immunology , Clodronic Acid/metabolism , Female , Leukocyte Common Antigens/immunology , Liposomes/chemistry , Liposomes/metabolism , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/physiology , Monocytes/cytology , Stem Cells/cytology , Survival Rate , West Nile Fever/pathology , West Nile virus
9.
Am J Pathol ; 172(4): 993-1004, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18292235

ABSTRACT

Contact hypersensitivity is a T-cell-mediated response to a hapten. Exposing C57BL/6 mice to UV B radiation systemically suppresses both primary and secondary contact hypersensitivity responses. The effects of UVB on in vivo T-cell responses during UVB-induced immunosuppression are unknown. We show here that UVB exposure, before contact sensitization, inhibits the expansion of effector CD4+ and CD8+ T cells in skin-draining lymph nodes and reduces the number of CD4+ and IFN-gamma+ CD8+ T cells infiltrating challenged ear skin. In the absence of UVB, at 10 weeks after initial hapten exposure, the ear skin of sensitized mice was infiltrated by dermal effector memory CD8+ T cells at the site of challenge. However, if mice were previously exposed to UVB, this cell population was absent, suggesting an impaired development of peripheral memory T cells. This finding occurred in the absence of UVB-induced regulatory CD4+ T cells and did not involve prostaglandin E2, suggesting that the importance of these two factors in mediating or initiating UVB-induced immunosuppression is dependent on UVB dose. Together these data indicate that in vivo T-cell responses are prone to immunoregulation by UVB, including a novel effect on both the activated T-cell pool size and the development of memory T cells in peripheral compartments.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/radiation effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Immunity/radiation effects , Immunologic Memory/radiation effects , Ultraviolet Rays , Animals , Cell Movement/radiation effects , Cell Proliferation , Dermatitis, Contact/immunology , Ear/pathology , Ear/radiation effects , Female , Green Fluorescent Proteins/metabolism , Interferon-gamma/biosynthesis , Leukocytes/pathology , Leukocytes/radiation effects , Lymph Nodes/immunology , Lymph Nodes/radiation effects , Lymphocyte Activation/radiation effects , Mice , Mice, Inbred C57BL , Phenotype , Skin/pathology , Skin/radiation effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects
10.
Photochem Photobiol ; 84(1): 35-46, 2008.
Article in English | MEDLINE | ID: mdl-18173699

ABSTRACT

Both the UVB and UVA wavebands within sunlight are immunosuppressive. This article reviews the relationship between wavebands and dose in UV-induced immunosuppression mainly concentrating on responses in humans. It also contrasts the effects of UVB and UVA on cellular changes involved in immunosuppression. Over physiological sunlight doses to which humans can be exposed during routine daily living or recreational pursuits, both UVA and UVB suppress immunity. While there is a linear dose relationship with UVB commencing at doses less than half of what is required to cause sunburn, UVA has a bell-shaped dose response over the range to which humans can be realistically exposed. At doses too low for either waveband to be suppressive, interactions between UVA and UVB augment each other, enabling immunosuppression to occur. At doses beyond where UVA is immunosuppressive, it still contributes to sunlight-induced immunosuppression via this interaction with UVB. While there is little research comparing the mechanisms by which UVB, UVA and their interactions can cause immunosuppression, it is likely that different chromophores and early molecular events are involved. There is evidence that both wavebands disrupt antigen presentation and effect T cell responses. Different individuals are likely to have different immunomodulatory responses to sunlight.


Subject(s)
Immune System/immunology , Immune System/radiation effects , Sunlight , Animals , Dose-Response Relationship, Radiation , Humans , Hypersensitivity/immunology , Neoplasms/immunology , Signal Transduction/immunology , Signal Transduction/radiation effects
11.
Immunol Cell Biol ; 85(1): 33-42, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17146465

ABSTRACT

With the recent emergence of the flavivirus, West Nile virus (WNV), in particular, the New York strain of Lineage I WNV in North America in 1999, there has been a significant increase in activity in neurotropic flavivirus research. These viruses cause encephalitis that can result in permanent neurological sequelae or death. Attempts to develop vaccines have made progress, but have been variably successful, despite considerable commercial underwriting. Thus, the discovery of ways and means to combat disease is no less urgent. As such, most recent work has been directed towards dissecting and understanding the pathogenesis of disease, as a way of informing possible approaches to abrogation or amelioration of illness. Whether inherent to flaviviruses or because humans are incidental, dead-end hosts, it is clear that these viruses interact with their human hosts in extremely complex ways. This occurs from the cellular level, at which infection must be established to produce disease, to its interaction with the adaptive immune response, which may result in its eradication, with or without immunopathological and consequent neurological sequelae. As human proximity to and contact with flavivirus insect vectors and amplifying hosts cannot practically be eliminated, our understanding of the pathogenesis of flavivirus-induced diseases, especially with regard to possible targets for treatment, is imperative.


Subject(s)
Central Nervous System/virology , Flavivirus Infections/immunology , Flavivirus Infections/pathology , Animals , Antibody Formation , Central Nervous System/immunology , Flavivirus/immunology , Flavivirus/pathogenicity , Flavivirus Infections/transmission , Humans , Immunity, Cellular , Immunity, Innate , Mice , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...