Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Mil Med ; 186(Suppl 1): 364-369, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33499483

ABSTRACT

INTRODUCTION: Negative Pressure Wound Therapy (NPWT) is a procedure used for nonhealing wounds. In NPWT, a special sealed dressing of large cell foam (>400 µm) or gauze is connected to a pump. Most commonly, negative pressures between -10 and -125 millimeters of mercury (mm Hg) are used. The mechanism of healing is unknown but maybe attributable to removal of the exudate and bacteria, and the stimulation of tissue repair through microdeformation. Reticulated foams with micron-size open cells, Capillary Suction Devices (CSD; 100 to 5 µm) exert capillary suction between 10 and 70 mm of Hg with a multilayered foam dressing. MATERIALS AND METHODS: Yorkshire pigs received 5 surgical excision wounds, 3 cm2, on each side of the back. The wounds were covered with a NPWT dressing (110 mm Hg negative pressure by a pump), CSD with capillary suctions of 30 mm Hg (CSD-30) and 70 mm Hg (CSD-70), and a conventional gauze dressing. The wounds were measured on day 2, and then every 4-5 days thereafter; the total fluid collected by the various dressing over time. RESULTS: By post-wound day 20, the wounds treated with CSD-70 and NPWT were 100% closed while the wounds treated with CSD-30 and gauze were 65% and 45%, respectively. This indicated comparable wound closure efficacies for CSD-70 and NPWT. The average total fluid uptake measured in grams dry weight were similar for CSD-70 and NPWT, 36 and 38 g, respectively, while the values were 24 g for CSD-30 and 12 g for gauze. However, the maximum fluid uptake observed at day 2 indicated that CSD-70 and CSD 30, 24 and 14 g, respectively, were superior to NPWT and gauze 12 and 7 g, respectively. CONCLUSION: This data indicate comparable wound closure efficacies for CSD-70 and NPWT. It is felt that CSD is an effective, safe, and lower cost alternative to vacuum-assisted NPWT.


Subject(s)
Negative-Pressure Wound Therapy , Animals , Bandages , Suction , Swine , Wound Healing
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
5.
Stem Cell Res ; 9(2): 124-34, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22705496

ABSTRACT

DNA double-strand breaks (DSBs) in embryonic stem (ES) cells are repaired primarily by homologous recombination (HR). The mechanism by which HR is regulated in these cells, however, remains enigmatic. To gain insight into such regulatory mechanisms, we have asked how protein levels of Rad51, a key component of HR, are controlled in mouse ES cells and mouse embryo fibroblasts (MEFs). The Rad51 protein level is about 15-fold higher in ES cells than in MEFs. The level of Rad51 mRNA, however, is only ~2-fold higher, indicating that the differences in mRNA levels due to rates of transcription or mRNA stability are not sufficient to account for the large difference in the abundance of Rad51 protein. Comparison of Rad51 half-lives between ES cells and MEFs also did not explain the elevated level of Rad51 protein in the ES cells. A comparative assessment of the Rad51 translation level demonstrated that it is translated with much greater efficacy in ES cells than in MEFs. To determine whether this high level of translation in ES cells is a general phenomenon in these cells or whether it is a characteristic of specific proteins, such as those involved with recombination and cell cycle progression, we compared mechanisms that regulate the level of Pcna in ES cells with those that regulate Rad51. The half-life of Pcna and its rate of synthesis were considerably different from those of Rad51 in ES cells, demonstrating that regulation of Rad51 abundance cannot be generalized to other ES cell proteins and not to proteins involved in DNA replication and cell cycle control. Finally, we show that only a small proportion of the abundant Rad51 protein population is activated under basal conditions in ES cells and recruited to DNA DSBs and/or stalled replication forks.


Subject(s)
Embryonic Stem Cells/metabolism , Rad51 Recombinase/metabolism , Animals , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA Replication/genetics , E2F Transcription Factors/metabolism , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Mice , Proliferating Cell Nuclear Antigen/biosynthesis , Protein Biosynthesis , Protein Stability , Rad51 Recombinase/genetics
6.
J Invest Dermatol ; 132(9): 2255-62, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22572817

ABSTRACT

The melanocortin 1 receptor (MC1R), a G(s) protein-coupled receptor, has an important role in human pigmentation. We investigated the regulation of expression and activity of the MC1R in primary human melanocyte cultures. Human ß-defensin 3 (HBD3) acted as an antagonist for MC1R, inhibiting the α-melanocortin (α-melanocyte-stimulating hormone (α-MSH))-induced increase in the activities of adenylate cyclase and tyrosinase, the rate-limiting enzyme for melanogenesis. α-Melanocortin and forskolin, which activate adenylate cyclase, and 12-O-tetradecanoylphorbol-13-acetate, which activates protein kinase C, increased, whereas exposure to UV radiation reduced, MC1R gene and membrane protein expression. Brief treatment with α-MSH resulted in MC1R desensitization, whereas continuous treatment up to 3 hours caused a steady rise in cAMP, suggesting receptor recycling. Pretreatment with agouti signaling protein or HBD3 prohibited responsiveness to α-MSH, but not forskolin, suggesting receptor desensitization by these antagonists. Melanocytes from different donors expressed different levels of the G protein-coupled receptor kinases (GRKs) 2, 3, 5, and 6, as well as ß-arrestin 1. Therefore, in addition to the MC1R genotype, regulation of MC1R expression and activity is expected to affect human pigmentation and the responses to UV.


Subject(s)
Agouti Signaling Protein/pharmacology , Melanocortins/pharmacology , Melanocytes/drug effects , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/antagonists & inhibitors , alpha-MSH/pharmacology , beta-Defensins/pharmacology , Adenylyl Cyclases/metabolism , Arrestins/biosynthesis , Cells, Cultured , Colforsin/pharmacology , G-Protein-Coupled Receptor Kinases/biosynthesis , Humans , Melanocytes/metabolism , Melanocytes/radiation effects , Monophenol Monooxygenase/metabolism , Protein Kinase C/metabolism , Receptor, Melanocortin, Type 1/biosynthesis , Skin Pigmentation/drug effects , Skin Pigmentation/physiology , Skin Pigmentation/radiation effects , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Ultraviolet Rays , beta-Arrestin 1 , beta-Arrestins
7.
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
8.
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
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.
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
11.
J Burn Care Res ; 31(1): 1-12, 2010.
Article in English | MEDLINE | ID: mdl-20061831

ABSTRACT

Psychological stress has a high incidence after burn injury, therefore, anxiolytic drugs are often prescribed. Unfortunately, to date, no burn study has investigated the effects of anxiolytic drugs on the ability to fight infection. This study was undertaken to determine if psychological stress, anxiety-modulating drugs, or both, alter survival following an infection. On day 0, 7-week-old male C57Bl/6 mice either received a 15% full-thickness flame burn or were sham treated (anesthesia and shaved), whereas controls received no treatment. Mice received midazolam (1 mg/kg intraperitoneally) or saline daily and were stressed by exposure to rat in a guinea pig cage or placed in an empty cage for 1 hour a day, beginning on postburn day 1. For the survival experiments, mice either received bacteria after 2 or 8 consecutive days of predator exposure and drug treatment, which continued daily for 7 days after inoculation. In a separate set of experiments, after eight daily injections of midazolam, mice were given lipopolysaccharide, bacteria, or saline and were killed 12 hours later. Mice that received midazolam had improved survival rates when compared with their saline-treated counterparts, and the protective effect was more significant the more days they received the drug. For most of the cytokines, the bacteria-induced increase was significantly attenuated by midazolam as was the amount of bacteria in the liver. The protective effect seems to be independent of the drug's anxiolytic activity as there were no significant differences in survival between the predator-stressed and the nonstressed mice. The mechanisms responsible for the protective effect remain to be elucidated.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Burns/psychology , Midazolam/therapeutic use , Pseudomonas Infections/therapy , Pseudomonas aeruginosa , Stress, Psychological/drug therapy , Animals , Burns/microbiology , Burns/therapy , Male , Mice , Mice, Inbred C57BL , Pseudomonas Infections/etiology , Pseudomonas Infections/psychology , Rats , Rats, Long-Evans , Stress, Psychological/complications , Stress, Psychological/microbiology
12.
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
13.
Genesis ; 47(6): 423-31, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19415629

ABSTRACT

Transforming growth factor beta1 (TGFbeta1) is a multifunctional growth factor involved in wound healing, tissue fibrosis, and in the pathogenesis of many syndromic diseases (e.g., Marfan syndrome, Camurati-Engelmann disease) and muscular, neurological, ophthalmic, cardiovascular and immunological disorders, and cancer. Since the generation of Tgfb1 knockout mice, there has been extraordinary progress in understanding its physiological and pathophysiological function. Here, we report the generation of a conditional knockout allele for Tgfb1 in which its exon 6 is flanked with LoxP sites. As proof of principle, we crossed these mice to LckCre transgenic mice and specifically disrupted Tgfb1 in T cells. The results indicate that T-cell-produced TGFbeta1 is required for normal in vivo regulation of peripheral T-cell activation, maintenance of T-cell homeostasis, and suppression of autoimmunity.


Subject(s)
Exons/genetics , Gene Targeting/methods , T-Lymphocytes/metabolism , Transforming Growth Factor beta1/genetics , Alleles , Animals , Cell Count , Female , Flow Cytometry , Gene Expression Profiling , Homeostasis/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Spleen/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/metabolism , Transforming Growth Factor beta1/physiology
14.
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
15.
J Cell Biol ; 180(6): 1073-86, 2008 Mar 24.
Article in English | MEDLINE | ID: mdl-18347065

ABSTRACT

The S-phase checkpoint activated at replication forks coordinates DNA replication when forks stall because of DNA damage or low deoxyribonucleotide triphosphate pools. We explore the involvement of replication forks in coordinating the S-phase checkpoint using dun1Delta cells that have a defect in the number of stalled forks formed from early origins and are dependent on the DNA damage Chk1p pathway for survival when replication is stalled. We show that providing additional origins activated in early S phase and establishing a paused fork at a replication fork pause site restores S-phase checkpoint signaling to chk1Delta dun1Delta cells and relieves the reliance on the DNA damage checkpoint pathway. Origin licensing and activation are controlled by the cyclin-Cdk complexes. Thus, oncogene-mediated deregulation of cyclins in the early stages of cancer development could contribute to genomic instability through a deficiency in the forks required to establish the S-phase checkpoint.


Subject(s)
Cell Division/genetics , DNA Damage/genetics , DNA Replication/genetics , Genes, cdc/physiology , S Phase/genetics , Saccharomyces cerevisiae/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Checkpoint Kinase 1 , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/metabolism , Gene Expression Regulation, Fungal/genetics , Genomic Instability/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Time Factors
16.
Clin Immunol ; 127(2): 206-13, 2008 May.
Article in English | MEDLINE | ID: mdl-18308639

ABSTRACT

TGFbeta1 is considered to be required for peripheral maintenance of CD4(+)CD25(+)FOXP3(+) T(reg) cells. However, we demonstrate no reduction in the percentage of such T cells in the spleens and thymi of Tgfb1(-/-) mice. Although putative T(reg) cells, characterized as CD4(+)CD25(+)FOXP3(+)CD62L(+) T cells, are increased in Tgfb1(-/-) mice, they may be inadequate to control activated T cells since the ratio of activated T cells:putative T(reg) cells is several-fold higher in Tgfb1(-/-) mice than in control mice. We further show that whereas Tgfb1(-/-) mice that express a chicken OVA-specific TCR transgene (DO11.10) have an increase in putative T(reg) cells, there are no detectable CD4(+)CD25(+) T cells in the spleens of DO11.10 Rag1(-/-) mice suggesting that T(reg)-cell generation is self-antigen dependent regardless of whether they express Tgfb1. Finally, we demonstrate that Tgfb1(-/-) T cells remain responsive to the suppressive effect of TGFbeta1 in vitro. These data suggest that TGFbeta1 is required for the regulatory function of T(reg) cells to prevent activation of T cells and autoimmunity.


Subject(s)
Forkhead Transcription Factors/immunology , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Autoimmunity/immunology , Cell Proliferation , Flow Cytometry , L-Selectin/immunology , Lectins, C-Type , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Phenotype , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , Thymus Gland/cytology , Thymus Gland/immunology , Transforming Growth Factor beta1/genetics
17.
Neuroimmunomodulation ; 14(6): 287-96, 2007.
Article in English | MEDLINE | ID: mdl-18287810

ABSTRACT

OBJECTIVE: Current evidence supports the conclusion that prolactin (PRL) is not an obligate immunoregulatory hormone and influences the immune system predominantly during stress conditions. In this study, we examined the impact of PRL on the psychogenic stress-induced responses of myeloid cells. METHODS: Seven-week-old PRL+/- (normal) and PRL-/- (deficient) mice were exposed to a predator for 1 h/day on 3 consecutive days. Another group of PRL-deficient mice received either 1 pituitary graft (hyperprolactinemic) or sham surgery at 5 weeks of age, while PRL-normal mice only received sham surgery. Two weeks later, these mice were also subjected to predator exposure. One day after the last predator exposure session, all mice were killed and the bone marrow and blood harvested. RESULTS: Significant differences in the myeloid cells between PRL-normal and PRL-deficient mice only occurred in stressed conditions. The median serum corticosterone levels were consistently higher in PRL-deficient mice. The implantation of a pituitary graft lowered the corticosterone levels to those observed in PRL-normal mice. The absolute number of immature neutrophils as well as the numbers of granulocyte macrophage, monocyte/macrophage and granulocyte colonies were significantly higher in the stressed PRL-deficient mice; however, only the increased number of immature neutrophils was reversed by pituitary grafting. CONCLUSIONS: Our findings support previous observations that PRL influences myeloid cells of the bone marrow most profoundly in stressed conditions. However, the mechanism by which PRL influences bone marrow myeloid cells during stress cannot be explained solely by its effect on serum corticosterone.


Subject(s)
Bone Marrow Cells/physiology , Chemokines/blood , Glucocorticoids/blood , Myeloid Cells/physiology , Prolactin/metabolism , Stress, Psychological/physiopathology , Animals , Behavior, Animal , Flow Cytometry , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neuroimmunomodulation/physiology , Neutrophils/metabolism , Prolactin/genetics
18.
Mol Cancer Ther ; 5(12): 3181-90, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172422

ABSTRACT

Prostatic adenocarcinomas depend on androgen for growth and survival. First line treatment of disseminated disease exploits this dependence by specifically targeting androgen receptor function. Clinical evidence has shown that androgen receptor is reactivated in recurrent tumors despite the continuance of androgen deprivation therapy. Several factors have been shown to restore androgen receptor activity under these conditions, including somatic mutation of the androgen receptor ligand-binding domain. We have shown previously that select tumor-derived mutants of the androgen receptor are receptive to activation by bisphenol A (BPA), an endocrine-disrupting compound that is leached from polycarbonate plastics and epoxy resins into the human food supply. Moreover, we have shown that BPA can promote cell cycle progression in cultured prostate cancer cells under conditions of androgen deprivation. Here, we challenged the effect of BPA on the therapeutic response in a xenograft model system of prostate cancer containing the endogenous BPA-responsive AR-T877A mutant protein. We show that after androgen deprivation, BPA enhanced both cellular proliferation rates and tumor growth. These effects were mediated, at least in part, through androgen receptor activity, as prostate-specific antigen levels rose with accelerated kinetics in BPA-exposed animals. Thus, at levels relevant to human exposure, BPA can modulate tumor cell growth and advance biochemical recurrence in tumors expressing the AR-T877A mutation.


Subject(s)
Adenocarcinoma/drug therapy , Androgen Receptor Antagonists , Phenols/pharmacology , Prostatic Neoplasms/drug therapy , Adenocarcinoma/pathology , Androgens/metabolism , Animals , Benzhydryl Compounds , Cell Growth Processes/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
19.
Lab Invest ; 86(10): 1008-19, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16865088

ABSTRACT

To investigate whether the multifocal inflammatory disease in TGFbeta1-deficient mice is caused by self-antigen (self-Ag)-specific autoreactive T cells, or whether it is caused by antigen independent, spontaneous hyperactivation of T cells, we have generated Tgfb1(-/-) and Tgfb1(-/-) Rag1(-/-) mice expressing the chicken OVA-specific TCR transgene (DO11.10). On a Rag1-sufficient background, Tgfb1(-/-) DO11.10 mice develop a milder inflammation than do Tgfb1(-/-) mice, and their T cells display a less activated phenotype. The lower level of activation correlates with the expression of hybrid TCR (transgenic TCRbeta and endogenous TCRalpha), which could recognize self-Ag and undergo activation. In the complete absence of self-Ag recognition (Tgfb1(-/-) DO11.10 Rag1(-/-) mice) inflammation and T-cell activation are eliminated, demonstrating that self-Ag recognition is required for the hyper-responsiveness of TGFbeta1-deficient T cells. Thus, TGFbeta1 is required for the prevention of autoimmune disease through its ability to control the activation of autoreactive T cells to self-Ag.


Subject(s)
Autoimmunity/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta1/immunology , Animals , Autoantigens/immunology , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Mice, Transgenic
20.
Cancer Res ; 65(11): 4568-77, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15930274

ABSTRACT

The liver exhibits an exquisitely controlled cell cycle, wherein hepatocytes are maintained in quiescence until stimulated to proliferate. The retinoblastoma tumor suppressor, pRB, plays a central role in proliferative control by inhibiting inappropriate cell cycle entry. In many cases, liver cancer arises due to aberrant cycles of proliferation, and correspondingly, pRB is functionally inactivated in the majority of hepatocellular carcinomas. Therefore, to determine how pRB loss may provide conditions permissive for deregulated hepatocyte proliferation, we investigated the consequence of somatic pRB inactivation in murine liver. We show that liver-specific pRB loss results in E2F target gene deregulation and elevated cell cycle progression during post-natal growth. However, in adult livers, E2F targets are repressed and hepatocytes become quiescent independent of pRB, suggesting that other factors may compensate for pRB loss. Therefore, to probe the consequences of acute pRB inactivation in livers of adult mice, we gave adenoviral-Cre by i.v. injection. We show that acute pRB loss is sufficient to elicit E2F target gene expression and cell cycle entry in adult liver, demonstrating a critical role for pRB in maintaining hepatocyte quiescence. Finally, we show that liver-specific pRB loss results in the development of nuclear pleomorphism associated with elevated ploidy that is evident in adult mice harboring both acute and chronic pRB loss. Together, these results show the crucial role played by pRB in maintaining hepatocyte quiescence and ploidy in adult liver in vivo and underscore the critical importance of delineating the consequences of acute pRB loss in adult animals.


Subject(s)
Liver/physiology , Ploidies , Retinoblastoma Protein/deficiency , Animals , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Growth Processes/genetics , DNA-Binding Proteins/genetics , E2F Transcription Factors , Gene Silencing , Genes, Retinoblastoma/genetics , Hepatocytes/cytology , Hepatocytes/physiology , Liver/cytology , Liver/metabolism , Mice , Mice, Knockout , Retinoblastoma Protein/genetics , Retinoblastoma Protein/physiology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...