Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Biomedicines ; 11(2)2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36831131

ABSTRACT

BACKGROUND: Cancer treatment has many side effects; therefore, more efficient treatments are needed. Mesenchymal stem cells (MSC) have immunoregulatory properties, tumor site migration and can be genetically modified. Some proteins, such as soluble TRAIL (sTRAIL) and interleukin-12 (IL-12), have shown antitumoral potential, thus its combination in solid tumors could increase their activity. MATERIALS AND METHODS: Lentiviral transduction of bone marrow MSC with green fluorescent protein (GFP) and transgenes (sTRAIL and IL-12) was confirmed by fluorescence microscopy and Western blot. Soluble TRAIL levels were quantified by ELISA. Lymphoma L5178Y cells express a reporter gene (GFP/mCherry), and TRAIL receptor (DR5). RESULTS: An in vivo model showed that combined treatment with MSC expressing sTRAIL+IL-12 or IL-12 alone significantly reduced tumor volume and increased survival in BALB/c mice (p < 0.05) with only one application. However, at the histological level, only MSC expressing IL-12 reduced tumor cell infiltration significantly in the right gastrocnemius compared with the control group (p < 0.05). It presented less tissue dysplasia confirmed by fluorescence and hematoxylin-eosin dye; nevertheless, treatment not inhibited hepatic metastasis. CONCLUSIONS: MSC expressing IL-12, is or combination with BM-MSC expressing sTRAIL represents an antitumor strategy for lymphoma tumors since they increase survival and reduce tumor development. However, the combination did not show significative additive effect. The localized application did not inhibit metastasis but reduced morphological alterations of tissue associated with liver metastasis.

2.
Mol Med Rep ; 25(6)2022 06.
Article in English | MEDLINE | ID: mdl-35485288

ABSTRACT

As the understanding of cancer grows, new therapies have been proposed to improve the well-known limitations of current therapies, whose efficiency relies mostly on early detection, surgery and chemotherapy. Mesenchymal stem cells (MSCs) have been introduced as a promissory and effective therapy. This fact is due to several useful features of MSCs, such as their accessibility and easy culture and expansion in vitro, and their remarkable ability for 'homing' towards tumors, allowing MSCs to exert their anticancer effects directly into tumors. Additionally, MSCs offer the practicability of being genetically engineered to carry anticancer genes, increasing their specificity and efficacy for fighting tumors. In the present study, the antitumoral efficacy and post-implant survival of mice bearing lymphomas implanted intratumorally were determined using mouse bone marrow-derived (BM)-MSCs transduced with soluble TRAIL (sTRAIL), full length TRAIL (flTRAIL), or interferon ß (IFNß), naïve BM-MSCs, or combinations of these. The percentage of surviving mice was determined once all not-implanted mice succumbed. It was found that the percentage of surviving mice implanted with the combination of MSCs-sTRAIL and MSCs-IFN-ß was 62.5%. Lymphoma model achieved 100% fatality in the non-treated group by day 41. On the other hand, the percentage of surviving mice implanted with MSCs-sTRAIL was 50% and with MSCs-INFß 25%. All the aforementioned differences were statistically significant (P<0.05). In conclusion, all implants exhibited tumor size reduction, growth delay, or apparent tumor clearance. MSCs proved to be effective anti-lymphoma agents; additionally, the combination of soluble TRAIL and IFN-ß resulted in the most effective antitumor and life enlarging treatment, showing an additive antitumoral effect compared with individual treatments.


Subject(s)
Lymphoma , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Hypertrophy , Interferon-beta/genetics , Lymphoma/genetics , Lymphoma/therapy , Mice
3.
Acta Trop ; 222: 106041, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34252383

ABSTRACT

Cutaneous leishmaniasis is the most common form of leishmaniasis in humans. The disease is caused by several species, such as Leishmania mexicana, a protozoa parasite. Several major risk factors are associated with this disease, including poverty, poor housing, inadequate domestic hygiene, malnutrition, mobility, and occupational exposure. Solar radiation (UVB) has not been considered a risk factor because there is no scientific evidence demonstrating a correlation with increased susceptibility to cutaneous leishmaniasis. In this study, the shaved skin of the back of C57BL/6 mice was irradiated with 24.2 mJ/cm2 of UVB. A delayed-type hypersensitivity (DTH) reaction was used to assess UV-induced immune suppression. Skin lesions were quantitated, and parasite burden and the presence of anti-Leishmania mexicana antibodies in serum and germinal centers in draining lymph nodes were determined. We found an increased in the lesion size and parasitic load in UVB-irradiated mice compared to the WT mice and B lymphocyte activation in draining lymph nodes and increased IgG1 production. Our results show an important role of UVB-induced suppression in cutaneous leishmaniasis through local production of IL-10 and systemic IgG1antibodies. This is the first study that demonstrates the effects of UVB radiation on cutaneous leishmaniasis by Leishmania mexicana.


Subject(s)
Immunosuppression Therapy , Leishmaniasis, Cutaneous , Skin/radiation effects , Ultraviolet Rays , Animals , Leishmania mexicana , Leishmaniasis, Cutaneous/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin/parasitology
4.
Exp Dermatol ; 25(7): 537-43, 2016 07.
Article in English | MEDLINE | ID: mdl-26914366

ABSTRACT

Psoralen and UVA (PUVA) has immunosuppressive and proapoptotic effects, which are thought to be responsible alone or in combination for its therapeutic efficacy. However, the molecular mechanism by which PUVA mediates its effects is not well understood. Activation of the serotonin (5-hydroxytryptamine, 5-HT) pathway has been suggested to be involved in the modulation of T-cell responses and found to mediate UVB-induced immune suppression. In particular, the activation of the 5-HT2A receptor has been proposed as one mechanism responsible for UV-induced immune suppression. We therefore hypothesized that 5-HT may play a role in PUVA-induced effects. The model of systemic suppression of delayed-type hypersensitivity (DTH) to Candida albicans was used to study immune function after exposure of C3H and KIT(W) (-Sh/W-Sh) mice to a minimal inflammatory dose of topical PUVA. The intra-peritoneal injection of the 5-HT2 receptor antagonist ketanserin or cyproheptadine or an anti-5-HT antibody immediately before PUVA exposure entirely abrogated suppression of DTH but had no significant effect on inflammation, as measured by swelling and cellular infiltration of the skin, and apoptosis as determined by the number of sunburn cells in C3H mice. Importantly, the systemic injection of 5-HT recapitulated PUVA immune suppression of DTH but did not induce inflammation or apoptosis in the skin. KIT(W) (-Sh/W-Sh) mice (exhibiting myelopoietic abnormalities, including lack of 5-HT-containing mast cells) were resistant to PUVA-induced suppression of DTH but not local skin swelling. Thus, this points towards a crucial role of 5-HT signalling in PUVA-induced immune suppression but not inflammation or apoptosis in situ in the skin.


Subject(s)
Hypersensitivity, Delayed/metabolism , PUVA Therapy , Serotonin/metabolism , Animals , Apoptosis , Female , Hypersensitivity, Delayed/drug therapy , Inflammation/metabolism , Mast Cells/physiology , Mice, Inbred C3H
5.
Exp Dermatol ; 24(7): 491-6, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25776520

ABSTRACT

Dermal mast cells protect the skin from inflammatory effects of ultraviolet (UV) radiation and are required for UV-induced immune suppression. We sought to determine a potential mechanistic role of mast cells in reducing the sensitivity to UV radiation (i.e. phototolerance induction) through photohardening. We administered single UV exposures as well as a chronic UV irradiation regime to mast cell-deficient Kit(W-Sh/W-Sh) mice and their controls. The chronic irradiation protocol was similar to that given for prophylaxis in certain photodermatoses in humans. Compared to controls, UV-exposed Kit(W-Sh/W-Sh) mice were more susceptible to epidermal hyperplasia and dermal oedema which was linked to blood vessel dilation. Unexpectedly, Kit(W-Sh/W-Sh) mice exhibited an excessive scratching behaviour following broadband UVB plus UVA or solar simulated UV irradiation at doses far below their minimal skin-swelling dose. Protection from this UV-induced scratching phenotype was dependent on mast cells, as engraftment of bone marrow-derived cultured mast cells abated it entirely. Kit(W-Sh/W-Sh) mice were entirely resistant to phototolerance induction by photohardening treatment. Compared to controls, these mice also showed reduced numbers of regulatory T cells and neutrophils in the skin 24 h after UV irradiation. While it is well known that mast cell-deficient mice are resistant to UV-induced immune suppression, we have discovered that they are prone to develop photo-itch and are more susceptible to UV-induced epidermal hyperplasia and skin oedema.


Subject(s)
Mast Cells/immunology , Mast Cells/radiation effects , Skin/immunology , Skin/radiation effects , Ultraviolet Rays/adverse effects , Animals , Edema/etiology , Edema/immunology , Hyperplasia , Immune Tolerance/radiation effects , Mast Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophils/immunology , Neutrophils/radiation effects , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , Pruritus/etiology , Pruritus/immunology , Pruritus/physiopathology , Skin/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects , Vasodilation/immunology , Vasodilation/radiation effects
6.
J Leukoc Biol ; 95(1): 139-48, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24009177

ABSTRACT

The UVB (290-320 nm) radiation in sunlight is responsible for inducing skin cancer. Exposure to UV radiation is also immunosuppressive, and the systemic immune suppression induced by UV is a well-recognized risk factor for cancer induction. As UVB radiation is absorbed within the upper layers of the skin, indirect mechanisms must play a role in activating systemic immune suppression. One prominent example is mast cell migration, which from the skin to the draining LN is an essential step in the cascade of events leading to immune suppression. What triggers mast cell migration is not entirely clear. Here, we tested the hypothesis that PAF, a lipid mediator of inflammation produced by the skin in response to UV exposure, is involved. Mast cell-deficient mice (Kit(W-sh/W-sh)) are resistant to the suppressive effect of UV radiation, and reconstituting mast cell-deficient mice with normal bone marrow-derived mast cells restores susceptibility to immunosuppression. However, when mast cells from PAFR-/- mice were used, the reconstituted mice were not susceptible to the suppressive effects of UV. Furthermore, PAFR-/- mice showed impaired UV-induced mast cell migration when compared with WT mice. Finally, injecting PAF into WT mice mimicked the effect of UV irradiation and induced mast cell migration but not in PAFR-/- mice. Our findings indicate that PAFR binding induces mast cells to migrate from the skin to the LNs, where they mediate immune suppression.


Subject(s)
Chemotaxis/genetics , Chemotaxis/immunology , Mast Cells/immunology , Mast Cells/metabolism , Platelet Activating Factor/genetics , Animals , Chemotaxis/drug effects , Chemotaxis/radiation effects , Female , Gene Expression , Gene Expression Regulation , Immunosuppression Therapy , Mast Cells/drug effects , Mast Cells/radiation effects , Mice , Mice, Knockout , Platelet Activating Factor/metabolism , Platelet Activating Factor/pharmacology , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Ultraviolet Rays
7.
BMC Cancer ; 11: 36, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21269511

ABSTRACT

BACKGROUND: The incidence of non-Hodgkin's lymphoma has increased over recent years. The exact etiology of lymphoma remains unknown. Ultraviolet light exposure has been associated with the development of internal lymphoid malignancies and some reports suggest that it may play a role in the development of lymphoma in humans. Here we describe the characterization and progression of lymphoma in p53 heterozygous mice exposed to UVB irradiation. METHODS: UVB-irradiated p53+/- mice developed enlargement of the spleen. Isolated spleen cells were transplanted into Rag deficient hosts. The UV-induced tumor cells were analyzed by flow cytometry. The tumor cells were tagged with GFP to study their metastatic potential. SKY and karyotypic analysis were carried out for the detection of chromosomal abnormalities. Functional assays included in vitro class switch recombination assay, immunoglobulin rearrangement assay, as well as cytokine profiling. RESULTS: UVB-exposed mice showed enlargement of the spleen and lymph nodes. Cells transplanted into Rag deficient mice developed aggressive tumors that infiltrated the lymph nodes, the spleen and the bone marrow. The tumor cells did not grow in immune competent syngeneic C57Bl/6 mice yet showed a modest growth in UV-irradiated B6 mice. Phenotypic analysis of these tumor cells revealed these cells are positive for B cell markers CD19+, CD5+, B220+, IgM+ and negative for T cell, NK or dendritic cell markers. The UV-induced tumor cells underwent robust in vitro immunoglobulin class switch recombination in response to lipopolysaccharide. Cytogenetic analysis revealed a t(14;19) translocation and trisomy of chromosome 6. These tumor cells secret IL-10, which can promote tumor growth and cause systemic immunosuppression. CONCLUSION: UV-irradiated p53+/- mice developed lymphoid tumors that corresponded to a mature B cell lymphoma. Our results suggest that an indirect mechanism is involved in the development of internal tumors after chronic exposure to UV light. The induction of B cell lymphoma in UV-irradiated p53 heterozygous mice may provide a useful model for lymphoma development in humans.


Subject(s)
Genes, p53 , Haploinsufficiency , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/genetics , Ultraviolet Rays , Animals , Chromosome Aberrations , Cytokines/biosynthesis , Interleukin-10/biosynthesis , Interleukin-10/metabolism , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
J Immunol ; 186(1): 25-31, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21098222

ABSTRACT

The most prevalent cancer diagnosed in the world is sunlight-induced skin cancer. In addition to being a complete carcinogen, UV radiation, the causative agent of skin cancer, induces immune suppression. Because UV-induced immune suppression is a well-recognized risk factor for skin cancer induction, it is crucial to understand the mechanisms underlying UV-induced immune suppression. Mast cells, which have recently emerged as immune regulatory cells, are particularly important in UV-induced immune suppression. UV exposure does not induce immune suppression in mast cell-deficient mice. We report that UV irradiation blocks germinal center (GC) formation, Ab secretion, and T follicular helper (Tfh) cell function, in part by altering the expression of transcription factors BCL-6 and BLIMP-1. No suppression of GC formation, Tfh cell IL-21 expression, or Ab secretion was observed in UV-irradiated mast cell-deficient (Kit(W-sh/W-sh)) mice. When mast cell-deficient mice were reconstituted with wild type mast cells, immune suppression was restored. Reconstituting the mast cell-deficient mice with bone marrow-derived mast cells from IL-10-deficient mice failed to restore the ability of UV radiation to suppress GC formation. Our findings demonstrate a function for mast cells, suppression of Tfh cell production, GC formation, and Ab production in vivo.


Subject(s)
Cell Differentiation/immunology , Germinal Center/cytology , Germinal Center/immunology , Growth Inhibitors/physiology , Interleukin-10/physiology , Lymphocyte Activation/immunology , Mast Cells/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibody Formation/radiation effects , Bone Marrow Transplantation/immunology , Cell Differentiation/radiation effects , Germinal Center/radiation effects , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukins/antagonists & inhibitors , Interleukins/biosynthesis , Interleukins/radiation effects , Lymphocyte Activation/genetics , Mast Cells/metabolism , Mast Cells/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-6/biosynthesis , Proto-Oncogene Proteins c-bcl-6/radiation effects , Proto-Oncogene Proteins c-kit/genetics , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/radiation effects , Ultraviolet Rays
9.
Toxicol Sci ; 112(1): 144-52, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726579

ABSTRACT

Applying jet propulsion-8 (JP-8) jet fuel to the skin of mice induces immune suppression. Applying JP-8 to the skin of mice suppresses T-cell-mediated immune reactions including, contact hypersensitivity (CHS) delayed-type hypersensitivity and T-cell proliferation. Because dermal mast cells play an important immune regulatory role in vivo, we tested the hypothesis that mast cells mediate jet fuel-induced immune suppression. When we applied JP-8 to the skin of mast cell deficient mice CHS was not suppressed. Reconstituting mast cell deficient mice with wild-type bone marrow derived mast cells (mast cell "knock-in mice") restored JP-8-induced immune suppression. When, however, mast cells from prostaglandin E(2) (PGE(2))-deficient mice were used, the ability of JP-8 to suppress CHS was not restored, indicating that mast cell-derived PGE(2) was activating immune suppression. Examining the density of mast cells in the skin and lymph nodes of JP-8-treated mice indicated that jet fuel treatment caused an initial increase in mast cell density in the skin, followed by increased numbers of mast cells in the subcutaneous space and then in draining lymph nodes. Applying JP-8 to the skin increased mast cell expression of CXCR4, and increased the expression of CXCL12 by draining lymph node cells. Because CXCL12 is a chemoattractant for CXCR4+ mast cells, we treated JP-8-treated mice with AMD3100, a CXCR4 antagonist. AMD3100 blocked the mobilization of mast cells to the draining lymph node and inhibited JP-8-induced immune suppression. Our findings demonstrate the importance of mast cells in mediating jet fuel-induced immune suppression.


Subject(s)
Hydrocarbons/toxicity , Mast Cells/drug effects , Skin/drug effects , Animals , Chemokines/metabolism , Dinoprostone/genetics , Dinoprostone/physiology , Lymph Nodes/cytology , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Skin/cytology , Skin/immunology
10.
Toxicol Sci ; 108(1): 100-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19095747

ABSTRACT

Applying jet fuel (JP-8) to the skin of mice induces immune suppression. JP-8-treated keratinocytes secrete prostaglandin E(2), which is essential for activating immune suppressive pathways. The molecular pathway leading to the upregulation of the enzyme that controls prostaglandin synthesis, cyclooxygenase (COX)-2, is unclear. Because JP-8 activates oxidative stress and because reactive oxygen species (ROS) turn on nuclear factor kappa B (NF-kappabeta), which regulates the activity of COX-2, we asked if JP-8-induced ROS and NF-kappabeta contributes to COX-2 upregulation and immune suppression in vivo. JP-8 induced the production of ROS in keratinocytes as measured with the ROS indicator dye, aminophenyl fluorescein. Fluorescence was diminished in JP-8-treated keratinocytes overexpressing catalase or superoxide dismutase (SOD) genes. JP-8-induced COX-2 expression was also reduced to background in the catalase and SOD transfected cells, or in cultures treated with N-acetylcysteine (NAC). When NAC was injected into JP-8-treated mice, dermal COX-2 expression, and JP-8-induced immune suppression was inhibited. Because ROS activates NF-kappabeta, we asked if this transcriptional activator played a role in the enhanced COX-2 expression and JP-8-induced immune suppression. When JP-8-treated mice, or JP-8-treated keratinocytes were treated with a selective NF-kappabeta inhibitor, parthenolide, COX-2 expression, and immune suppression were abrogated. Similarly, when JP-8-treated keratinocytes were treated with small interfering RNA specific for the p65 subunit of NF-kappabeta, COX-2 upregulation was blocked. These data indicate that ROS and NF-kappabeta are activated by JP-8, and these pathways are involved in COX-2 expression and the induction of immune suppression by jet fuel.


Subject(s)
Hydrocarbons/toxicity , Immune Tolerance/drug effects , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Administration, Cutaneous , Analysis of Variance , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Gene Expression/drug effects , Hydrocarbons/pharmacology , Mice , NF-kappa B/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Small Interfering/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Sesquiterpenes/pharmacology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factor RelA
11.
Vet Immunol Immunopathol ; 124(3-4): 220-9, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18482772

ABSTRACT

Armadillos are apparently important reservoirs of Mycobacterium leprae and an animal model for human leprosy, whose immune system has been poorly studied. We aimed at characterizing the armadillo's langerhans cells (LC) using epidermal sheets instead of tissue sections, since the latter restrict analysis only to cut-traversed cells. Epidermal sheets by providing an en face view, are particularly convenient to evaluate dendritic morphology (cells are complete), spatial distribution (regular vs. clustered), and frequency (cell number/tissue area). Lack of anti-armadillo antibodies was overcome using LC-restricted ATPase staining, allowing assessment of cell frequency, cell size, and dendrites extension. Average LC frequency in four animals was 528 LC/mm(2), showing a rather uniform non-clustered distribution, which increased towards the animal's head, while cell size increased towards the tail; without overt differences between sexes. The screening of antibodies to human DC (MHC-II, CD 1a, langerin, CD86) in armadillo epidermal sheets, revealed positive cells with prominent dendritic morphology only with MHC-II and CD86. This allowed us to test DC mobilization from epidermis into dermis under topical oxazolone stimulation, a finding that was corroborated using whole skin conventional sections. We hope that the characterization of armadillo's LC will incite studies of leprosy and immunity in this animal model.


Subject(s)
Armadillos/anatomy & histology , Epidermal Cells , Langerhans Cells/cytology , ADP-ribosyl Cyclase 1/immunology , Adenosine Triphosphatases/biosynthesis , Adjuvants, Immunologic/pharmacology , Animals , Antibodies/immunology , Armadillos/immunology , Biopsy/veterinary , Cross Reactions , Dendritic Cells/drug effects , Dendritic Cells/immunology , Epidermis/enzymology , Epidermis/immunology , Female , HLA-DR Antigens/immunology , Immunohistochemistry/veterinary , Langerhans Cells/enzymology , Langerhans Cells/immunology , Male , Oxazolone/pharmacology
12.
J Immunol ; 180(7): 4648-55, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18354188

ABSTRACT

The UV radiation in sunlight is the primary cause of skin cancer. UV is also immunosuppressive and numerous studies have shown that UV-induced immune suppression is a major risk factor for skin cancer induction. Previous studies demonstrated that dermal mast cells play a critical role in the induction of immune suppression. Mast cell-deficient mice are resistant to the immunosuppressive effects of UV radiation, and UV-induced immune suppression can be restored by injecting bone marrow-derived mast cells into the skin of mast cell- deficient mice. The exact process however, by which mast cells contribute to immune suppression, is not known. In this study, we show that one of the first steps in the induction of immune suppression is mast cell migration from the skin to the draining lymph nodes. UV exposure, in a dose-dependent manner, causes a significant increase in lymph node mast cell numbers. When GFP(+) skin was grafted onto mast cell-deficient mice, we found that GFP(+) mast cells preferentially migrated into the lymph nodes draining the skin. The mast cells migrated primarily to the B cell areas of the draining nodes. Mast cells express CXCR4(+) and UV exposure up-regulated the expression of its ligand CXCL12 by lymph node B cells. Treating UV-irradiated mice with a CXCR4 antagonist blocked mast cell migration and abrogated UV-induced immune suppression. Our findings indicate that UV-induced mast cell migration to draining lymph nodes, mediated by CXCR4 interacting with CXCL12, represents a key early step in UV-induced immune suppression.


Subject(s)
Cell Movement/immunology , Cell Movement/radiation effects , Lymph Nodes/cytology , Lymph Nodes/immunology , Mast Cells/immunology , Skin/cytology , Skin/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Chemokine CXCL12/immunology , Chemokine CXCL12/metabolism , Immune Tolerance/immunology , Immune Tolerance/radiation effects , Lymph Nodes/metabolism , Lymph Nodes/radiation effects , Mast Cells/cytology , Mast Cells/radiation effects , Mice , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Skin/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...