Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Pigment Cell Melanoma Res ; 27(4): 601-10, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24730569

ABSTRACT

Activation of the melanocortin 1 receptor (MC1R) by α-melanocortin (α-MSH) stimulates eumelanin synthesis and enhances repair of ultraviolet radiation (UV)-induced DNA damage. We report on the DNA damage response (DDR) of human melanocytes to UV and its enhancement by α-MSH. α-MSH up-regulated the levels of XPC, the enzyme that recognizes DNA damage sites, enhanced the UV-induced phosphorylation of the DNA damage sensors ataxia telangiectasia and Rad3-related (ATR) and ataxia telangiectasia mutated (ATM) and their respect-ive substrates checkpoint kinases 1 and 2, and increased phosphorylated H2AX (γH2AX) formation. These effects required functional MC1R and were absent in melanocytes expressing loss of function (LOF) MC1R. The levels of wild-type p53-induced phosphatase 1 (Wip1), which dephosphorylates γH2AX, correlated inversely with γH2AX. We propose that α-MSH increases UV-induced γH2AX to facilitate formation of DNA repair complexes and repair of DNA photoproducts, and LOF of MC1R compromises the DDR and genomic stability of melanocytes.


Subject(s)
DNA Damage , DNA Repair/radiation effects , Genomic Instability/radiation effects , Melanocytes/metabolism , Receptor, Melanocortin, Type 1/metabolism , Ultraviolet Rays/adverse effects , Adult , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Ataxia Telangiectasia Mutated Proteins/genetics , Cells, Cultured , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Female , Histones/genetics , Histones/metabolism , Humans , Infant , Infant, Newborn , Male , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 2C , Receptor, Melanocortin, Type 1/genetics , Up-Regulation/radiation effects , alpha-MSH/genetics , alpha-MSH/metabolism
2.
J Immunotoxicol ; 11(2): 148-59, 2014.
Article in English | MEDLINE | ID: mdl-23895378

ABSTRACT

Tungstate (WO²â»4) has been identified as a ground water contaminant at military firing ranges and can be absorbed by ingestion. In this study, C57BL6 mice were exposed to sodium tungstate (Na2WO4·2H2O) (0, 2, 62.5, 125, and 200 mg/kg/day) in their drinking water for an initial 28-day screen and in a one-generation (one-gen) model. Twenty-four hours prior to euthanasia, mice were intraperitoneally injected with Staphylococcal enterotoxin B (SEB) (20 µg/mouse) or saline as controls. After euthanasia, splenocytes and blood were collected and stained with lymphocyte and/or myeloid immunophenotyping panels and analyzed by flow cytometry. In the 28-day and one-gen exposure, statistically significant reductions were observed in the quantities of activated cytotoxic T-cells (TCTL; CD3(+)CD8(+)CD71(+)) and helper T-cells (TH; CD3(+)CD4(+)CD71(+)) from spleens of SEB-treated mice. In the 28-day exposures, CD71(+) TCTL cells were 12.87 ± 2.05% (SE) in the 0 tungstate (control) group compared to 4.44 ± 1.42% in the 200 mg/kg/day (p < 0.001) group. TH cells were 4.85 ± 1.23% in controls and 2.76 ± 0.51% in the 200 mg/kg/day (p < 0.003) group. In the one-gen exposures, TCTL cells were 7.98 ± 0.49% and 6.33 ± 0.49% for P and F1 mice after 0 mg/kg/day tungstate vs 1.58 ± 0.23% and 2.52 ± 0.25% after 200 mg/kg/day of tungstate (p < 0.001). Similarly, TH cells were reduced to 6.21 ± 0.39% and 7.20 ± 0.76%, respectively, for the 0 mg/kg/day P and F1 mice, and 2.28 ± 0.41% and 2.85 ± 0.53%, respectively, for the 200 mg/kg/day tungstate P and F1 groups (p < 0.001). In delayed-type hypersensitivity Type IV experiments, tungstate exposure prior to primary and secondary antigen challenge significantly reduced footpad swelling at 20 and 200 mg/kg/day. These data indicate that exposure to tungstate can result in immune suppression that may, in turn, reduce host defense against pathogens.


Subject(s)
Adaptive Immunity/drug effects , Tungsten Compounds/pharmacology , Administration, Oral , Animals , Enterotoxins , Female , Hypersensitivity, Delayed/immunology , Immunophenotyping , Interferon-gamma/biosynthesis , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology
3.
Inflammation ; 35(1): 259-70, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21400122

ABSTRACT

Burn patients requiring hospitalization are often treated for anxiety with benzodiazepines (BDZs). Benzodiazepines are reported to influence immune system function. Immune system alterations are a major cause of burn-induced mortality. We wanted to determine whether the BDZ, midazolam given daily at an anxiolytic dose, had any influence on the burn injury-induced inflammatory response in the blood and wound. Mice received a 15% total body surface area flame burn and received either midazolam 1 mg/kg i.p. or saline 0.1 ml daily. Blood and skin wounds were harvested 24 h after injection on post-burn day 2, 3, 7, or 8. Mice treated with midazolam had significantly lower serum IL-1ß (p=0.002), TNF-α (p=0.002), IL-6 (p=0.016), IL-10 (p=0.009), and TGF-ß (p=0.004) than saline-treated mice, with little impact on serum chemokine levels. In the wound, TNF-α and IL-10 were the only cytokines significantly influenced by the drug, being lower (p=0.018) and higher (p=0.006), respectively. The chemokines in the wound influenced significantly by midazolam were MIP-1α, MIP-1ß, and MIP-2 while MCP-1 and KC were not. There were more inflammatory cells at the burn wound margin in midazolam-treated mice on post-burn day 3. Although serum nitrate/nitrite was significantly increased by midazolam (p=0.03), both eNOS and iNOS mRNA expression in the wound were similar to the saline group. We found that midazolam given daily after burn injury significantly influenced the inflammatory response. The clinical implications of these findings on wound healing and shock following burn injury, especially larger burns, deserve further investigation.


Subject(s)
Burns/immunology , Inflammation/metabolism , Midazolam/pharmacology , Animals , Chemokine CCL2/blood , Chemokine CCL3/blood , Chemokine CCL4/blood , Chemokine CXCL2/blood , Inflammation/pathology , Interleukin-10/blood , Interleukin-1beta/blood , Interleukin-6/blood , Male , Mice , Mice, Inbred C57BL , Midazolam/administration & dosage , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III/genetics , RNA, Messenger/biosynthesis , Skin/pathology , Transforming Growth Factor beta/blood , Tumor Necrosis Factor-alpha/blood , Wound Healing/physiology
4.
Nucleic Acids Res ; 39(17): 7465-76, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21653549

ABSTRACT

The human DEK gene is frequently overexpressed and sometimes amplified in human cancer. Consistent with oncogenic functions, Dek knockout mice are partially resistant to chemically induced papilloma formation. Additionally, DEK knockdown in vitro sensitizes cancer cells to DNA damaging agents and induces cell death via p53-dependent and -independent mechanisms. Here we report that DEK is important for DNA double-strand break repair. DEK depletion in human cancer cell lines and xenografts was sufficient to induce a DNA damage response as assessed by detection of γH2AX and FANCD2. Phosphorylation of H2AX was accompanied by contrasting activation and suppression, respectively, of the ATM and DNA-PK pathways. Similar DNA damage responses were observed in primary Dek knockout mouse embryonic fibroblasts (MEFs), along with increased levels of DNA damage and exaggerated induction of senescence in response to genotoxic stress. Importantly, Dek knockout MEFs exhibited distinct defects in non-homologous end joining (NHEJ) when compared to their wild-type counterparts. Taken together, the data demonstrate new molecular links between DEK and DNA damage response signaling pathways, and suggest that DEK contributes to DNA repair.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , DNA Breaks, Double-Stranded , DNA Repair , Oncogene Proteins/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Mice , Mice, Knockout , Mice, Nude , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Poly-ADP-Ribose Binding Proteins , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism
5.
Shock ; 36(2): 149-55, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21558983

ABSTRACT

Burn induces myeloid-derived suppressor cells (MDSCs), a heterogeneous population of immature polymorphonuclear neutrophils (PMNs) and monocytes, which protect against infection. Previous work from our laboratory demonstrated that inflammatory monocytes (iMos) were the major MDSC source of TNF-α in the postburn spleen, and we hypothesized that they were also the major source of postburn IL-10. To test this hypothesis, we examined cytokine production by postburn CCR2 knockout (KO) mice, which have fewer iMos than burn wild-type (WT) splenocytes, but equal numbers of PMNs and F4/80 macrophages. Using cell sorting and/or intracellular cytokine techniques, we examined IL-10 production by postburn PMNs and iMos. Finally, we compared IL-10 production by postburn PMNs and iMos with culture-derived MDSCs. Splenocytes from postburn CCR2 KO mice produced less IL-6 and TNF-α than WT burn splenocytes in response to LPS, but KO and WT burn splenocytes produced equal amounts of IL-10 in response to peptidoglycan. Depletion of PMNs from postburn splenocytes led to reductions in IL-10 and increases in IL-6 and TNF-α in response to peptidoglycan, but not in response to LPS. Sorting or intracellular cytokine techniques gave consistent results: Burn PMNs made more IL-10 than sham PMNs and also more IL-10 than burn or sham iMos. Polymorphonuclear neutrophil and iMos subpopulations from culture-derived MDSCs produced the same cytokine profiles in response to LPS and peptidoglycan as did the PMNs and iMos from postburn spleens: PMNs made IL-10, whereas iMos made IL-6. Finally, LPS-induced mortality of burn mice was made worse by anti-Gr-1 depletion of all PMNs and 66% of iMos from burn mice. This suggests that PMNs play a primarily anti-inflammatory role in vitro and in vivo.


Subject(s)
Burns/immunology , Interleukin-10/metabolism , Macrophages/immunology , Monocytes/immunology , Neutrophils/immunology , Receptors, CCR2/metabolism , Spleen/metabolism , Animals , Burns/physiopathology , Flow Cytometry , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Myeloid Cells/metabolism , Neutrophils/metabolism , Receptors, CCR2/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
J Invest Dermatol ; 131(6): 1226-33, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21326292

ABSTRACT

Modification of skin complexion coloration has traditionally been accomplished by interruption or attenuation of melanogenesis and/or melanosome transfer. Post-transfer modification of pigmented melanosomes provides an attractive and distinct avenue of modulating skin pigmentation. The processing of melanosomes during keratinocyte (KC) terminal differentiation and the degradative variability observed between light and dark skin (LS and DS) remains enigmatic. To evaluate this, we developed a model system to investigate the loss of fluorescently labeled and isolated melanosomes by cultured human KCs. The extent of melanosome loss has been qualitatively assessed using transmission electron microscopy and indirect immunofluorescence with confocal microscopy, and quantitatively assessed using flow cytometry analysis. Results show that melanosomes are incorporated into the cytoplasm of both light and dark keratinocytes (LKCs and DKCs) and trafficked to a perinuclear region. Within 48 hours, confocal microscopy images suggest that LKCs display accelerated melanosome loss. This time-dependent decrease in carboxyfluorescein diacetate (CFDA) fluorescence was then quantitatively analyzed using flow cytometry. Consistent with the results of the confocal analysis, over a 48-hour time frame, LKCs appear to lose melanosomes more efficiently than DKCs. These experiments show that melanosomes are more rapidly lost in KCs derived from LS as opposed to DS.


Subject(s)
Epidermal Cells , Keratinocytes/ultrastructure , Melanosomes/ultrastructure , Skin Pigmentation , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Microscopy, Confocal , Microscopy, Electron, Transmission
7.
DNA Repair (Amst) ; 10(4): 445-51, 2011 Apr 03.
Article in English | MEDLINE | ID: mdl-21315663

ABSTRACT

Accumulation of mutations in embryonic stem (ES) cells would be detrimental to an embryo derived from these cells, and would adversely affect multiple organ systems and tissue types. ES cells have evolved multiple mechanisms to preserve genomic integrity that extend beyond those found in differentiated cell types. The present study queried whether mismatch repair (MMR) and base-excision repair (BER) may play a role in the maintenance of murine ES cell genomes. The MMR proteins Msh2 and Msh6 are highly elevated in mouse ES cells compared with mouse embryo fibroblasts (MEFs), as are Pms2 and Mlh1, albeit to a lesser extent. Cells transfected with an MMR reporter plasmid showed that MMR repair capacity is low in MEFs, but highly active in wildtype ES cells. As expected, an ES cell line defective in MMR was several-fold less effective in repair level than wildtype ES cells. Like proteins that participate in MMR, the level of proteins involved in BER was elevated in ES cells compared with MEFs. When BER activity was examined biochemically using a uracil-containing oligonucleotide template, repair activity was higher in ES cells compared with MEFs. The data are consistent with the suggestion that ES cells have multiple mechanisms, including highly active MMR and BER that preserve genetic integrity and minimize the accumulation of mutations.


Subject(s)
DNA Repair/physiology , Embryonic Stem Cells/metabolism , Animals , Cell Line , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Mice , Mice, Inbred C3H
8.
FASEB J ; 24(10): 3850-60, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20519635

ABSTRACT

The melanocortin 1 receptor gene is a main determinant of human pigmentation, and a melanoma susceptibility gene, because its variants that are strongly associated with red hair color increase melanoma risk. To test experimentally the association between melanocortin 1 receptor genotype and melanoma susceptibility, we compared the responses of primary human melanocyte cultures naturally expressing different melanocortin 1 receptor variants to α-melanocortin and ultraviolet radiation. We found that expression of 2 red hair variants abolished the response to α-melanocortin and its photoprotective effects, evidenced by lack of functional coupling of the receptor, and absence of reduction in ultraviolet radiation-induced hydrogen peroxide generation or enhancement of repair of DNA photoproducts, respectively. These variants had different heterozygous effects on receptor function. Microarray data confirmed the observed differences in responses of melanocytes with functional vs. nonfunctional receptor to α-melanocortin and ultraviolet radiation, and identified DNA repair and antioxidant genes that are modulated by α-melanocortin. Our findings highlight the molecular mechanisms by which the melanocortin 1 receptor genotype controls genomic stability of and the mutagenic effect of ultraviolet radiation on human melanocytes.


Subject(s)
Melanocytes/radiation effects , Receptor, Melanocortin, Type 1/genetics , Ultraviolet Rays , Cells, Cultured , Genotype , Humans
9.
Stem Cells Dev ; 19(11): 1699-711, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20446816

ABSTRACT

Embryonic stem (ES) cells give rise to all cell types of an organism. Since mutations at this embryonic stage would affect all cells and be detrimental to the overall health of an organism, robust mechanisms must exist to ensure that genomic integrity is maintained. To test this proposition, we compared the capacity of murine ES cells to repair DNA double-strand breaks with that of differentiated cells. Of the 2 major pathways that repair double-strand breaks, error-prone nonhomologous end joining (NHEJ) predominated in mouse embryonic fibroblasts, whereas the high fidelity homologous recombinational repair (HRR) predominated in ES cells. Microhomology-mediated end joining, an emerging repair pathway, persisted at low levels in all cell types examined. The levels of proteins involved in HRR and microhomology-mediated end joining were highly elevated in ES cells compared with mouse embryonic fibroblasts, whereas those for NHEJ were quite variable, with DNA Ligase IV expression low in ES cells. The half-life of DNA Ligase IV protein was also low in ES cells. Attempts to increase the abundance of DNA Ligase IV protein by overexpression or inhibition of its degradation, and thereby elevate NHEJ in ES cells, were unsuccessful. When ES cells were induced to differentiate, however, the level of DNA Ligase IV protein increased, as did the capacity to repair by NHEJ. The data suggest that preferential use of HRR rather than NHEJ may lend ES cells an additional layer of genomic protection and that the limited levels of DNA Ligase IV may account for the low level of NHEJ activity.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Embryonic Stem Cells/physiology , Recombination, Genetic , Animals , Antineoplastic Agents/pharmacology , Cells, Cultured , DNA Ligase ATP , DNA Ligases/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Fibroblasts/cytology , Fibroblasts/physiology , Mice , Mice, Inbred C57BL , Tretinoin/pharmacology
10.
Shock ; 34(5): 535-44, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20386495

ABSTRACT

Immature myeloid cells have been implicated as a source of postburn inflammation, and the appearance of these cells correlates with enhanced upregulation of hematopoiesis. The role of proliferative cells in postburn immune changes has not been directly tested. Gemcitabine, a ribonucleotide reductase inhibitor, has been shown to deplete proliferative immature myeloid cells in tumor models while sparing mature cells, leading to restored lymphocyte function and tumor regression. We treated burn mice at postburn day 6 (PBD6) with 120 mg/kg gemcitabine. On PBD8, splenocytes were taken and stimulated with LPS, peptidoglycan, or concanavalin A. The blood and spleen cell populations were enumerated by flow cytometry or automated cell counter. In addition, mice treated with gemcitabine were given LPS or infected with Pseudomonas aeruginosa at PBD8, and mortality was monitored. Gemcitabine depleted burn-induced polymorphonuclear leukocytes and inflammatory monocytes without affecting mature F4/80 macrophages. This was accompanied by reduced TNFα, IL-6, and IL-10 production by burn splenocytes. Burn splenocytes stimulated with mitogens exhibited increased nitric oxide production relative to sham mice. In vivo treatment of burn mice with gemcitabine blocked these burn-induced changes without damaging lymphocyte function. Treatment of burn mice with gemcitabine ameliorated burn-induced susceptibility to LPS and infiltration of polymorphonuclear leukocytes into the liver and lung. Finally, gemcitabine treatment blocked the protective effect of burn injury upon P. aeruginosa infection. Our report shows that proliferative cells are major drivers of postburn immune changes and provides evidence that implicates immature myeloid cells in these processes.


Subject(s)
Burns/immunology , Deoxycytidine/analogs & derivatives , Myeloid Cells/drug effects , Ribonucleotide Reductases/antagonists & inhibitors , Animals , Burns/drug therapy , Concanavalin A/pharmacology , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Evaluation, Preclinical , Interleukin-10/biosynthesis , Interleukin-6/biosynthesis , Leukocyte Count , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Macrophages/drug effects , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/physiology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Nitric Oxide/biosynthesis , Peptidoglycan/pharmacology , Pseudomonas Infections/complications , Spleen/immunology , Spleen/pathology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Gemcitabine
11.
J Immunotoxicol ; 7(3): 174-82, 2010.
Article in English | MEDLINE | ID: mdl-20178456

ABSTRACT

The potential for adverse health effects of using tungsten and its alloys in military munitions are an important concern to both civilians and the US military. The toxicological implications of exposure to tungsten, its alloys, and the soluble tungstate (Na(2)WO(4)) are currently under investigation. To examine tungstate toxicity, a series of experiments to determine its in vitro effects on cells of the immune system were performed. We identified alterations in isolated human peripheral blood lymphocytes (PBL) treated in vitro with sodium tungstate (0.01, 0.1, 1.0, and 10 mM). Analyses of apoptosis with annexin V and propidium iodide revealed a dose- and time-dependent increase in the quantity of cells in early apoptosis after tungstate exposure. Reductions in the number of cells entering into the cell cycle were also noted. Exposure of PBL to tungstate (1 mM) and Concanavalin A (ConA) for 72 h reduced the number of cells in S and G(2)/M phases of the cell cycle. There were alterations in the numbers of cells in G(0)/G(1), S, and G(2)/M phases of the cell cycle in long-term THP-1 (acute leukemic monocytes) cultures treated with tungstate (0.01, 0.1, 1.0, and 10 mM). Gel electrophoresis, silver staining, and LC-MS/MS showed the cytoplasmic presence of histone H1b and H1d after 72 h of tungstate exposure. The addition of tungstate to cultures resulted in significant reductions in the quantity of interleukin-10 (IL-10), tumor necrosis factor-alpha (TNF-alpha), and IL-6 produced by stimulated [CD3/CD28, ConA, or lipopolysaccharide (LPS)] and tungstate-treated lymphocytes. Taken together, these data indicate that tungstate increases apoptosis of PBL, alters cell cycle progression, reduces cytokine production, and therefore warrants further investigation.


Subject(s)
Apoptosis/drug effects , Cytokines/metabolism , Lymphocytes/drug effects , Tungsten Compounds/pharmacology , Adult , Apoptosis/immunology , Blood Circulation/immunology , Cell Cycle/drug effects , Cell Cycle/immunology , Cells, Cultured , Concanavalin A/immunology , Concanavalin A/metabolism , Cytokines/genetics , Cytoplasm/metabolism , Down-Regulation , Female , Histones/metabolism , Humans , Immunomodulation , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Middle Aged , Phenanthrenes/immunology , Phenanthrenes/metabolism
12.
Cancer Lett ; 290(2): 167-73, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-19796866

ABSTRACT

Resistance to chemotherapy is a major problem facing breast cancer patients. Cisplatin, a highly effective DNA-damaging drug, has shown only little success in breast cancer treatment. We are reporting that low nanomolar doses of bisphenol A (BPA) or estradiol antagonize cisplatin cytotoxicity in breast cancer cells, with their effects not mediated via classical estrogen receptors. Although both compounds increase the expression of Bcl-2, a Bcl-2 inhibitor completely blocked the protective effects of BPA while only partially affecting those of estradiol. Blockade of BPA and E2 actions should sensitize ER-negative breast tumors to anti-cancer drugs and allow for the inclusion of cisplatin in treatment regimens.


Subject(s)
Antineoplastic Agents/antagonists & inhibitors , Breast Neoplasms/drug therapy , Cisplatin/antagonists & inhibitors , Estradiol/pharmacology , Estrogens, Non-Steroidal/pharmacology , Phenols/pharmacology , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Benzhydryl Compounds , Blotting, Western , Cell Line, Tumor , Cell Separation , Cisplatin/toxicity , Drug Resistance, Neoplasm/drug effects , Estrogens/pharmacology , Female , Flow Cytometry , Humans , Reverse Transcriptase Polymerase Chain Reaction
13.
J Biol Chem ; 284(49): 34231-43, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19776016

ABSTRACT

Previously, we have shown that statistical synergism between amino acid variants in thyroglobulin (Tg) and specific HLA-DR3 pocket sequence signatures conferred a high risk for autoimmune thyroid disease (AITD). Therefore, we hypothesized that this statistical synergism mirrors a biochemical interaction between Tg peptides and HLA-DR3, which is key to the pathoetiology of AITD. To test this hypothesis, we designed a recombinant HLA-DR3 expression system that was used to express HLA-DR molecules harboring either AITD susceptibility or resistance DR pocket sequences. Next, we biochemically generated the potential Tg peptidic repertoire available to HLA-DR3 by separately treating 20 purified human thyroglobulin samples with cathepsins B, D, or L, lysosomal proteases that are involved in antigen processing and thyroid biology. Sequences of the cathepsin-generated peptides were then determined by matrix-assisted laser desorption ionization time-of-flight-mass spectroscopy, and algorithmic means were employed to identify putative AITD-susceptible HLA-DR3 binders. From four predicted peptides, we identified two novel peptides that bound strongly and specifically to both recombinant AITD-susceptible HLA-DR3 protein and HLA-DR3 molecules expressed on stably transfected cells. Intriguingly, the HLA-DR3-binding peptides we identified had a marked preference for the AITD-susceptibility DR signatures and not to those signatures that were AITD-protective. Structural analyses demonstrated the profound influence that the pocket signatures have on the interaction of HLA-DR molecules with Tg peptides. Our study suggests that interactions between Tg and discrete HLA-DR pocket signatures contribute to the initiation of AITD.


Subject(s)
Gene Expression Regulation , HLA-DR3 Antigen/metabolism , Recombinant Proteins/chemistry , Algorithms , Animals , Autoimmune Diseases , Cathepsins/chemistry , Cell Line , HeLa Cells , Histocompatibility Antigens Class II , Humans , Peptides/chemistry , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thyroglobulin/chemistry , Thyroid Diseases/immunology , Thyroid Gland/metabolism
14.
Carcinogenesis ; 30(8): 1298-304, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19443905

ABSTRACT

Resistance to chemotherapy is a major obstacle for successful treatment of breast cancer patients. Given that prolactin (PRL) acts as an anti-apoptotic/survival factor in the breast, we postulated that it antagonizes cytotoxicity by chemotherapeutic drugs. Treatment of breast cancer cells with PRL caused variable resistance to taxol, vinblastine, doxorubicin and cisplatin. PRL prevented cisplatin-induced G(2)/M cell cycle arrest and apoptosis. In the presence of PRL, significantly less cisplatin was bound to DNA, as determined by mass spectroscopy, and little DNA damage was seen by gamma-H2AX staining. PRL dramatically increased the activity of glutathione-S-transferase (GST), which sequesters cisplatin in the cytoplasm; this increase was abrogated by Jak and mitogen-activated protein kinase inhibitors. PRL upregulated the expression of the GSTmu, but not the pi, isozyme. A GST inhibitor abrogated antagonism of cisplatin cytotoxicity by PRL. In conclusion, PRL confers resistance against cisplatin by activating a detoxification enzyme, thereby reducing drug entry into the nucleus. These data provide a rational explanation for the ineffectiveness of cisplatin in breast cancer, which is characterized by high expression of both PRL and its receptor. Suppression of PRL production or blockade of its actions should benefit patients undergoing chemotherapy by allowing for lower drug doses and expanded drug options.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Glutathione Transferase/metabolism , Prolactin/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Nucleus/metabolism , Cytoplasm/metabolism , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Glutathione/metabolism , Histones/metabolism , Humans , Paclitaxel/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Cells, Cultured , Vinblastine/pharmacology
15.
Nat Cell Biol ; 11(4): 501-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287375

ABSTRACT

Impaired ribosome biogenesis is attributed to nucleolar disruption and diffusion of a subset of 60S ribosomal proteins, particularly ribosomal protein (rp)L11, into the nucleoplasm, where they inhibit MDM2, leading to p53 induction and cell-cycle arrest. Previously, we demonstrated that deletion of the 40S rpS6 gene in mouse liver prevents hepatocytes from re-entering the cell cycle after partial hepatectomy. Here, we show that this response leads to an increase in p53, which is recapitulated in culture by rpS6-siRNA treatment and rescued by the simultaneous depletion of p53. However, disruption of biogenesis of 40S ribosomes had no effect on nucleolar integrity, although p53 induction was mediated by rpL11, leading to the finding that the cell selectively upregulates the translation of mRNAs with a polypyrimidine tract at their 5'-transcriptional start site (5'-TOP mRNAs), including that encoding rpL11, on impairment of 40S ribosome biogenesis. Increased 5'-TOP mRNA translation takes place despite continued 60S ribosome biogenesis and a decrease in global translation. Thus, in proliferative human disorders involving hypomorphic mutations in 40S ribosomal proteins, specific targeting of rpL11 upregulation would spare other stress pathways that mediate the potential benefits of p53 induction.


Subject(s)
Cell Nucleolus/metabolism , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Humans , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Protein S6/deficiency , Ribosomal Protein S6/metabolism , Ribosomal Proteins/genetics , Transcription Initiation Site , Up-Regulation
16.
Mol Vis ; 14: 562-71, 2008 Mar 20.
Article in English | MEDLINE | ID: mdl-18385792

ABSTRACT

PURPOSE: To examine whether promiscuous Cre/LoxP recombination happens during gametogenesis in double transgenic mice carrying LoxP modified alleles and Cre transgene driven by tissue-specific promoter outside the gonads of adult mice. METHODS: Cre driver mice were crossbred with reporter mouse lines (e.g., ZEG and Rosa26R) to obtain Cre/ZEG and Cre/Rosa26R double transgenic mice. The frequency of promiscuous LoxP/Cre recombination was determined by the expression of second reporter genes in the offspring of double transgenic mice. RESULTS: The frequency of promiscuous LoxP/Cre recombination varied in different lines of Cre driver mice and in the sex of the same driver mice with higher penetrance in male than in female double transgenic mice. Polymerase chain reaction (PCR) and recombination analysis demonstrate that the recombination of floxed allele occurs during the transition from spermatogonia (diploid) to primary spermatocyte (tetraploid) in the testis. Thereby, target-floxed allele(s) may be ubiquitously ablated in experimental animals intended for tissue-specific gene deletion. CONCLUSIONS: Gametogenesis-associated recombination should always be examined in tissue-specific gene ablation studies.


Subject(s)
Alleles , Cornea/metabolism , Gametogenesis/genetics , Integrases , Recombination, Genetic , Animals , Epithelium, Corneal/metabolism , Female , Gene Deletion , Gene Expression , Gene Expression Regulation, Enzymologic , Genes, Reporter , Integrases/genetics , Keratin-12/genetics , Keratin-12/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Spermatogenesis/genetics
17.
Shock ; 28(6): 684-93, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17607156

ABSTRACT

Recent publications have demonstrated that human resident and inflammatory monocyte (IM) subpopulations have equivalents in rodents. The effect of thermal injury upon these subpopulations has not been studied. Mice were given a scald burn and killed on postburn days (PBDs) 2, 4, and 8. Bone marrow, blood, and spleen white cells were isolated, and the percentage of resident monocytes (CD11b LY6C), IMs (CD11b LY6C), and monocyte progenitors (macrophage-colony-forming unit [M-CFU]) were determined. The ability of each monocyte population to make TNF-alpha was determined by intracellular cytokine staining. Finally, the ability of sorted fractions from PBD 8 spleen to inhibit lymphocyte proliferation was performed. We noted that there was an increase in M-CFU in the blood and spleen at PBD 8, but the marrow only had a nonsignificant increase in M-CFU. All compartments showed a significant increase in the number of IMs by PBD 8, but no significant changes in resident monocytes were seen. In all compartments, IMs were a major source of TNF-alpha. The postburn increase in IMs and monocyte progenitors in the spleen was accompanied by an increase in the monocyte chemokine monocyte chemoattractant protein 1 and constitutively high levels of the progenitor chemokine stromal-derived factor 1alpha. After burn injury, mice deficient in the receptor for soluble TNF-alpha had equal levels of splenic M-CFU and monocytes, as did wild-type mice, suggesting that this cytokine is not essential for this effect. We conclude that in this model, IMs are a significant source of in vivo TNF-alpha.


Subject(s)
Burns/pathology , Inflammation/pathology , Monocytes/cytology , Animals , Bone Marrow Cells/immunology , Burns/blood , Burns/immunology , Cell Proliferation , Chemokine CCL2/metabolism , Cytokines/metabolism , Flow Cytometry , Humans , Inflammation/blood , Mice , Models, Biological , Monocytes/immunology , Monocytes/metabolism , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/immunology , Peptide Fragments/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Spleen/cytology , Spleen/immunology , Tumor Necrosis Factor Decoy Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Mol Cell Biol ; 27(17): 6127-39, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17606626

ABSTRACT

The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that mediates the toxic effects of its xenobiotic ligands and acts as an environmental checkpoint during the cell cycle. We expressed stably integrated, Tet-Off-regulated AHR variants in fibroblasts from AHR-null mice to further investigate the AHR role in cell cycle regulation. Ahr+/+ fibroblasts proliferated significantly faster than Ahr-/- fibroblasts did, and exposure to a prototypical AHR ligand or deletion of the ligand-binding domain did not change their proliferation rates, indicating that the AHR function in cell cycle was ligand independent. Growth-promoting genes, such as cyclin and cyclin-dependent kinase genes, were significantly down-regulated in Ahr-/- cells, whereas growth-arresting genes, such as the transforming growth factor beta1 (TGF-beta1) gene, extracellular matrix (ECM)-related genes, and cyclin-dependent kinase inhibitor genes, were up-regulated. Ahr-/- fibroblasts secreted significantly more TGF-beta1 into the culture medium than Ahr+/+ fibroblasts did, and Ahr-/- showed increased levels of activated Smad4 and TGF-beta1 mRNA. Inhibition of TGF-beta1 signaling by overexpression of Smad7 reversed the proliferative and gene expression phenotype of Ahr-/- fibroblasts. Changes in TGF-beta1 mRNA accumulation were due to stabilization resulting from decreased activity of TTP, the tristetraprolin RNA-binding protein responsible for mRNA destabilization through AU-rich motifs. These results show that the Ah receptor possesses interconnected intrinsic cellular functions, such as ECM formation, cell cycle control, and TGF-beta1 regulation, that are independent of activation by either exogenous or endogenous ligands and that may play a crucial role during tumorigenesis.


Subject(s)
Cell Cycle/physiology , Gene Expression Regulation , Receptors, Aryl Hydrocarbon/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Cell Proliferation , Cells, Cultured , ELAV Proteins , ELAV-Like Protein 1 , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Profiling , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Aryl Hydrocarbon/genetics , Smad7 Protein/genetics , Smad7 Protein/metabolism , Transforming Growth Factor beta1/genetics
19.
J Biol Chem ; 282(33): 23867-77, 2007 Aug 17.
Article in English | MEDLINE | ID: mdl-17556357

ABSTRACT

The retinoblastoma tumor suppressor (RB) is functionally inactivated in many human cancers. Classically, RB functions to repress E2F-mediated transcription and inhibit cell cycle progression. Consequently, RB ablation leads to loss of cell cycle control and aberrant expression of E2F target genes. Emerging evidence indicates a role for RB in maintenance of genomic stability. Here, mouse adult fibroblasts were utilized to demonstrate that aberrant DNA content in RB-deficient cells occurs concomitantly with an increase in levels and chromatin association of DNA replication factors. Furthermore, following exposure to nocodazole, RB-proficient cells arrest with 4 n DNA content, whereas RB-deficient cells bypass the mitotic block, continue DNA synthesis, and accumulate cells with higher ploidy and micronuclei. Under this condition, RB-deficient cells also retain high levels of tethered replication factors, MCM7 and PCNA, indicating that DNA replication occurs in these cells under nonpermissive conditions. Exogenous expression of replication factors Cdc6 or Cdt1 in RB-proficient cells does not recapitulate the RB-deficient cell phenotype. However, ectopic E2F expression in RB-proficient cells elevated ploidy and bypassed the response to nocodazole-induced cessation of DNA replication in a manner analogous to RB loss. Collectively, these results demonstrate that deregulated S phase control is a key mechanism by which RB-deficient cells acquire elevated ploidy.


Subject(s)
Cell Cycle Proteins/genetics , DNA Replication , Gene Expression Regulation , Nuclear Proteins/genetics , Ploidies , Retinoblastoma Protein/deficiency , Animals , Cells, Cultured , Chromatin/metabolism , DNA-Binding Proteins/genetics , E2F Transcription Factors/genetics , Fibroblasts/cytology , Mice , Minichromosome Maintenance Complex Component 7 , Proliferating Cell Nuclear Antigen/genetics , S Phase
20.
Cancer Biol Ther ; 6(4): 519-24, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17426441

ABSTRACT

Liposarcoma, a malignancy of adipose tissue, is the most common soft tissue sarcoma. Patients whose primary tumor cannot be resected or those who have developed metastasis, have poor prognosis since liposarcomas are highly resistant to chemotherapy. We recently generated a spontaneously immortalized cell line, named LS14, from a patient with metastatic liposarcoma. Our goal was to compare the responsiveness of LS14 and SW872 liposarcoma cells to anti-cancer drugs and explore mechanisms of chemoresistance. Using complementary assays for cell viability and number we found that SW872 cells responded robustly to relatively low concentrations of doxorubicin, cisplatin and vinblastine. This reduction in cell viability was due to apoptosis, as evident by phosphatidylserine exposure and caspase 3 cleavage. In contrast, only a high dose of doxorubicin or combination therapy effectively reduced LS14 cell viability and induced apoptosis. LS14 cells showed a higher expression of Bcl-2 and Bcl-xL, but a lower expression of survivin and Bax, than SW872 cells, suggesting that anti-apoptotic proteins contribute to chemoresistance in LS14 cells. Although LS14 cells did not form colonies in soft agar, they generated large tumors and metastases in SCID mice, establishing their tumorigenicity in vivo. In conclusion, LS14 cells are much more resistant to chemotherapy than SW872 cells, making them an excellent model for exploring the efficacy and mechanism of action of anti-cancer drugs in liposarcomas.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Line, Tumor/drug effects , Drug Resistance, Neoplasm , Liposarcoma , Models, Biological , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor/metabolism , Humans , Mice , Mice, SCID , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...