Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 299(3): 832-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11714866

ABSTRACT

We investigated the potential of arginine to reverse pathological changes in alcohol-induced liver injury. Four groups (six rats/group) of male Wistar rats were fed a fish oil-ethanol diet for 6 (group 2) or 8 (group 1) weeks. Rats in group 3 were fed fish oil-ethanol for 6 weeks, after which they were administered arginine with fish oil-ethanol for an additional 2 weeks. Rats in group 4 were fed fish oil-dextrose for 8 weeks. Liver samples were analyzed for histopathology, lipid peroxidation, cytochrome P4502E1 activity, nuclear factor-kappaB, and levels of messenger RNA for tumor necrosis factor-alpha, cyclooxygenase-2, and inducible nitric oxide synthase. Concentrations of endotoxin were measured in plasma. The most severe inflammation and fibrosis was detected in groups 1 and 2, as were the highest levels of endotoxin, lipid peroxidation, cytochrome P450 2E1 activity, activation of nuclear factor-kappaB, and mRNA levels for tumor necrosis factor-alpha, cyclooxygenase-2, and inducible nitric oxide synthase. Plasma nitric oxide was also increased as was nitrotyrosine in liver. After arginine was administered, there was marked improvement in the pathological changes accompanied by decreased levels of endotoxin, lipid peroxidation, activation of nuclear factor-kappaB, tumor necrosis factor-alpha, cyclooxygenase-2, inducible nitric oxide, and nitrotyrosine staining. The therapeutic effects of arginine are probably secondary to increased levels of nitric oxide but other effects of arginine cannot be excluded.


Subject(s)
Arginine/therapeutic use , Ethanol/toxicity , Liver Cirrhosis/prevention & control , Tyrosine/analogs & derivatives , Animals , Cyclooxygenase 2 , Disease Models, Animal , Down-Regulation , Drug Interactions , Endotoxins/metabolism , Humans , Inflammation/chemically induced , Inflammation/prevention & control , Isoenzymes/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Liver Cirrhosis, Alcoholic/prevention & control , Male , Membrane Proteins , NF-kappa B/metabolism , Nitric Oxide/blood , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism , Tyrosine/metabolism
2.
Transplantation ; 72(5): 948-53, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11571464

ABSTRACT

BACKGROUND: High-density oligoarray technology is a novel method for screening the expression of thousands of genes in a small tissue sample. Oligoarray analysis of genes expressed during human renal allograft rejection has not been reported previously. METHODS: Seven human renal allograft biopsies with histologic evidence of acute cellular rejection and three renal allograft biopsies without evidence of rejection (control) were analyzed for the expression of 6800 human genes using high-density oligoarrays (GeneChip, Affymetrix, Santa Clara, CA). Quantitative expression of gene transcripts was determined and a comparison analysis between acute rejection and control biopsy samples was performed. Up-regulation of a specific gene transcript during acute rejection was considered to be significant if transcript abundance increased fourfold or more relative to control biopsy samples. RESULTS: Comparison analysis revealed that between 32 and 219 gene transcripts are up-regulated (>fourfold) during acute rejection. Of these transcripts, only four (human monokine induced by interferon-gamma, T-cell receptor active beta-chain protein, interleukin-2 stimulated phosphoprotein, and RING4 (a transporter involved in antigen presentation)) were consistently up-regulated in each acute rejection sample relative to at least two of three control biopsy samples. Six other genes were up-regulated in six of seven acute rejection samples. These were interferon-stimulated growth factor-3, complement factor 3, nicotinamide N-methyltransferase, macrophage inflammatory protein-3beta, myeloid differentiation protein, and CD18. Only two gene transcripts were down-regulated in five of seven acute rejection samples. Significant up-regulation of cytotoxic T-cell effector molecules, previously reported as markers of acute renal rejection in humans, was not detected. CONCLUSIONS: High-density oligoarray technology is useful for screening gene expression in transplanted tissues undergoing acute rejection. Because this method does not rely on a priori knowledge of which genes are involved in acute rejection, it is likely to yield novel insights into the mechanisms and diagnosis of rejection.


Subject(s)
Gene Expression , Intercellular Signaling Peptides and Proteins , Kidney Transplantation/immunology , Oligonucleotide Array Sequence Analysis , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/genetics , Adult , Case-Control Studies , Chemokine CCL19 , Chemokine CXCL9 , Chemokines, CC/genetics , Chemokines, CXC/genetics , Female , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Kidney Transplantation/physiology , Male , Middle Aged , Phosphoproteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology
3.
Proc Natl Acad Sci U S A ; 98(10): 5550-5, 2001 May 08.
Article in English | MEDLINE | ID: mdl-11331784

ABSTRACT

Nox1, a homologue of gp91phox, the catalytic moiety of the superoxide (O(2)(-))-generating NADPH oxidase of phagocytes, causes increased O(2)(-) generation, increased mitotic rate, cell transformation, and tumorigenicity when expressed in NIH 3T3 fibroblasts. This study explores the role of reactive oxygen species (ROS) in regulating cell growth and transformation by Nox1. H(2)O(2) concentration increased approximately 10-fold in Nox1-expressing cells, compared with <2-fold increase in O(2)(-). When human catalase was expressed in Nox1-expressing cells, H(2)O(2) concentration decreased, and the cells reverted to a normal appearance, the growth rate normalized, and cells no longer produced tumors in athymic mice. A large number of genes, including many related to cell cycle, growth, and cancer (but unrelated to oxidative stress), were expressed in Nox1-expressing cells, and more than 60% of these returned to normal levels on coexpression of catalase. Thus, H(2)O(2) in low concentrations functions as an intracellular signal that triggers a genetic program related to cell growth.


Subject(s)
Cell Division/drug effects , Cell Transformation, Neoplastic/drug effects , Hydrogen Peroxide/pharmacology , NADH, NADPH Oxidoreductases/physiology , Animals , Base Sequence , Catalase/metabolism , Cell Division/physiology , Cell Line , DNA Primers , Mice , Mice, Nude , NADPH Oxidase 1 , Reverse Transcriptase Polymerase Chain Reaction
4.
Int J Dev Neurosci ; 16(5): 341-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9829170

ABSTRACT

G protein expression has been shown to be modulated by circulating plasma corticosterone in young animals. A loss of G protein expression regulation by corticosterone in the elderly could explain declines in the function of G protein-coupled receptors and their effective signal transduction processes in the nervous system found in normal and pathological aging. In this study, adrenalectomized 3- and 18-months-old female Fischer 344 rats were exposed to low, moderate or high levels of plasma corticosterone to determine the effect of this hormone on Gi protein expression in the hippocampus and frontal cortex. Basal Gi protein expression, assessed by Western blot analysis, did not vary across age in either brain region. Hippocampal Gi protein levels increased following moderate and high corticosterone administration in the 3-months old animals (125%; P < 0.05) but not in the 18-months old animals. In contrast, in the frontal cortex, Gi protein expression increased significantly in the 18-months-old group (93%; P < 0.05) following exposure to high concentrations of corticosterone. These results suggest that steroid hormones, specifically corticosterone, may differentially modulate neurotransmitter-G protein coupling in an age-dependent manner.


Subject(s)
Aging/metabolism , Corticosterone/pharmacology , Frontal Lobe/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/biosynthesis , Animals , Corticosterone/blood , Female , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Rats, Inbred F344
5.
Mamm Genome ; 9(7): 556-64, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9657854

ABSTRACT

Stannin is a protein that has been localized to trimethyltin-sensitive cell populations, and evidence suggests it plays a role in the toxic effects of organotins. In this study, we have isolated a mouse stannin genomic clone and have characterized the gene's intron-exon organization, promoter region, and chromosomal location. We have also isolated a partial human stannin cDNA clone and analyzed the open reading frame. The mouse genomic clone spans approximately 19 kb and consists of one intron and two exons. The splice site consensus sequence was maintained at all intron-exon junctions. Promoter analysis suggests that two putative promoter sites exist, each containing multiple regulatory elements and transcription factor-binding sites. Fluorescence in situ hybridization analysis localized stannin to mouse Chromosome (Chr) 16 at band A2. This region is homologous to the proximal region of human Chr 16 (16p13) to which stannin has been previously mapped. Sequence analysis revealed that the 264-bp open reading frame was identical between rat and mouse. The human sequence was 98% identical, with two amino acid substitutions near the c-terminal end of the peptide. These data suggest that stannin is highly conserved between species, and its unusual pattern of cellular expression may, in part, be explained via cell-specific promoters.


Subject(s)
Chromosome Mapping , Neuropeptides/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosomes, Human, Pair 16 , Cloning, Molecular , Conserved Sequence , DNA , Exons , Gene Library , Humans , Introns , Male , Mice , Molecular Sequence Data , Open Reading Frames , Promoter Regions, Genetic , RNA Splicing , Transcription, Genetic
6.
Hepatology ; 27(5): 1317-23, 1998 May.
Article in English | MEDLINE | ID: mdl-9581686

ABSTRACT

Increased hepatic oxidative stress with ethanol administration is hypothesized to be caused either by enhanced pro-oxidant production or decreased levels of antioxidants or both. We used the intragastric feeding rat model to assess the relationship between hepatic antioxidant enzymes and pathological liver injury in animals fed different dietary fats. Male Wistar rats (5 per group) were fed ethanol with either medium-chain triglycerides (MCTE), palm oil (PE), corn oil (CE), or fish oil (FE). Control animals were fed isocaloric amounts of dextrose instead of ethanol with the same diets. The following were evaluated in each group: liver pathology, lipid peroxidation, manganese superoxide dismutase (MnSOD) levels, copper-zinc SOD (CuZnSOD) levels, glutathione peroxidase (GPX) levels, and catalase (CAT) levels. All enzymes were evaluated using activity assays and immunoblots. Rats fed FE showed the most severe pathology (fatty liver, necrosis, and inflammation), those fed CE showed moderate changes, those fed PE showed fatty liver only, and those fed MCTE were normal. Parameters indicative of lipid peroxidation (conjugated dienes and thiobarbituric acid-reactive substances) were also greater in rat livers from animals fed the diets high in polyunsaturated fatty acids (CE and FE). CuZnSOD, GPX, and CAT activities showed an inverse correlation (r=-.92, P < .01) with severity of pathological injury, with the lowest levels for both enzymes found in FE-fed rats. Decreased enzyme activity in CE- and FE-fed rats was accompanied by similar decreases in immunoreactive protein. Ethanol administration did not cause significant decreases in enzyme activity in groups that showed no necroinflammatory changes (MCTE and PE). MnSOD activity showed no significant change in any ethanol-fed group. Our results show that decreases in CuZnSOD, GPX, and CAT occur in rats showing pathological liver injury and also having the highest levels of lipid peroxidation. These results suggest that feeding dietary substrates that enhance lipid peroxidation can exacerbate both ethanol-induced oxidative damage as well as necroinflammatory changes. The decrease in activity of antioxidant enzymes observed in animals fed diets high in polyunsaturated fatty acids and ethanol could possibly increase the susceptibility to oxidative damage and further contribute to ethanol-induced liver injury.


Subject(s)
Antioxidants/metabolism , Dietary Fats/metabolism , Hepatitis, Alcoholic/metabolism , Lipid Peroxides/metabolism , Animals , Catalase/metabolism , Corn Oil , Fish Oils , Glutathione Peroxidase/metabolism , Liver/enzymology , Male , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
7.
Neurochem Int ; 31(6): 801-15, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9413842

ABSTRACT

The cDNA encoding the protein stannin was isolated previously via subtractive hybridization, using differential expression after trimethyltin (TMT) intoxication, as a basis for isolating mRNA which may be expressed in TMT-sensitive cells. Initial characterization revealed a novel gene product which was differentially expressed in several tissues sensitive to TMT. In the current study, biochemical and molecular techniques were used to quantitate stannin expression at the cellular and subcellular levels. Northern blot analysis showed that the stannin 3.0 kb mRNA transcript was present, in decreasing amounts, in: spleen, hippocampus, neocortex, cerebellum, striatum, midbrain, kidney and lung. Liver, heart, skeletal muscle and testis showed no detectable expression of stannin mRNA. Immunoblot analysis using antipeptide antisera raised against stannin indicated a high level of expression in spleen, followed by brain and kidney. Stannin mRNA was present during early brain development and consolidated by post-natal day (PND) 20 to patterns and levels seen in adults. In situ hybridization studies showed widespread neuronal expression of stannin mRNA at PND 1, which shifted to a restricted pattern of expression in specific regions by PND 20. Stannin was partially purified from rodent brain and spleen using cation exchange, sizing and hydrophobic interaction chromatography. It behaved as a monomer throughout purification. Stannin was also expressed in a baculovirus system, using a series of constructs containing the entire cDNA, 1.0 kb of DNA flanking the open reading frame, and a 400 bp open reading frame minimal construct. While all constructs expressed stannin, the best expression was seen with the entire cDNA. Based on current findings, we suggest that stannin expression is necessary but not sufficient for TMT toxicity.


Subject(s)
Brain/drug effects , Neuropeptides/analysis , Organotin Compounds/pharmacology , Spleen/drug effects , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Baculoviridae/genetics , Blotting, Northern , Blotting, Western , Brain/metabolism , Gene Expression , Genetic Vectors , In Situ Hybridization , Male , Rats , Rats, Sprague-Dawley , Spleen/metabolism , Subcellular Fractions/chemistry , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
8.
Dig Dis Sci ; 42(10): 2106-10, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9365143

ABSTRACT

Nitric oxide (NO) is an important inhibitory neurotransmitter in the gut. Alterations in NO mediated responses have been described in diabetic animals. The presence of nitric oxide synthase (NOS) reflects the potential for NO synthesis and is found in neurons in the myenteric plexus. The aim of this study was to determine changes in nitric oxide synthase (NOS) expression in the myenteric plexus of the gastrointestinal tract of diabetic rats at three months of streptozotocin-induced diabetes, compared to age matched controls, using immunohistochemistry. Diabetic animals showed a decrease in NOS expression in the antrum, with 59.1 +/- 7.3% of neurons being positive for NOS in diabetes compared to 81.2 +/- 4.7% in controls (P < 0.05). NOS expression in duodenum, ileum, and colon of diabetic animals was not statistically different from controls. Decreased expression of NOS in antrum may contribute to altered gastric emptying observed in diabetics.


Subject(s)
Diabetes Mellitus, Experimental/enzymology , Myenteric Plexus/enzymology , Nitric Oxide Synthase/metabolism , Animals , Colon/enzymology , Duodenum/enzymology , Ileum/enzymology , Immunohistochemistry , Male , Neurons/enzymology , Pyloric Antrum/enzymology , Rats , Rats, Sprague-Dawley
9.
J Pharmacol Exp Ther ; 276(3): 1201-16, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8786553

ABSTRACT

Immortalized cell lines and primary neuronal cultures were used to characterize the selective toxicity of trimethyltin (TMT),triethyltin (TET) and tributyltin (TBT). TBT and TET were cytotoxic at similar concentrations in the immortalized cell lines tested; the 50% toxic concentration (TC50) was 1 to 11 microM. In contrast, immortalized cell lines varied considerably in their sensitivity to TMT, with sensitive cell lines (neuroblastomas, T-, B-cell lines) showing TC50 values of 2 to 8 microM, whereas insensitive cells (NIH-3T3 fibroblast, HTB-14 glioma, TC-7 kidney cells) had TC 50 values > 100 microM. Primary neuronal cell cultures were very sensitive to organotins (TC50 values, 1-10nM), and showed patterns of selective toxicity with respect to neuronal and glial cells. Because organotin toxicity evolves over 24 to 48 hr. we determined whether these compounds induced apoptosis in primary cultures. TMT increased (P < .05) the fraction of apoptotic cells 6 and 12 hr after treatment with TMT at TC50 concentrations. Prior studies suggested that a protein, stannin, was localized in cells sensitive to organotins. Stannin was expressed in several TMT-sensitive cell lines (PC12, T, B cells) and in primary neurons in culture. Stannin was absent in the resistant HTB-14 glioma cell line. The role of stannin in mediating TMT toxicity in primary cultures was investigated by blocking stannin expression with specific antisense oligonucleotides. Treatment of primary cultures with antisense oligonucleotides for 48 hr before and during TMT treatment significantly protected neurons from the neurotoxic and apoptotic effects of TMT. This effect was not observed with scrambled oligonucleotide controls. Thus, TMT may induce apoptosis in sensitive cells, which is partly mediated by stannin. Based on the available data we conclude that stannin expression is necessary, but not sufficient for TMT toxicity.


Subject(s)
Apoptosis/drug effects , Neuropeptides/pharmacology , Oligonucleotides, Antisense/pharmacology , Organotin Compounds/pharmacology , Animals , Blotting, Western , Cell Line , Dose-Response Relationship, Drug , Hippocampus/drug effects , Immunohistochemistry , In Vitro Techniques , Rats , Triethyltin Compounds/pharmacology , Trimethyltin Compounds/pharmacology
10.
Mol Pharmacol ; 47(4): 745-56, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7723735

ABSTRACT

Tau is a neuron-specific, microtubule-associated protein that forms paired helical filaments (PHFs) of Alzheimer's disease when aberrantly phosphorylated. We have attempted to elucidate the protein kinases and phosphatases that regulate tau phosphorylation. Incubation of rat, human, and rhesus monkey temporal neocortex slices with the phosphatase inhibitor okadaic acid induced epitopes of tau similar to those found in PHFs. Okadaic acid (1-20 microM) induced variant forms of tau at 60-68 kDa, which were recognized by the monoclonal antibodies Alz-50 (in humans only) and 5E2 and two polyclonal antipeptide antisera, OK-1 and OK-2. The phosphorylation-sensitive monoclonal antibody Tau-1 failed to recognize the slowest mobility forms of tau after okadaic acid treatment. FK-520 (1-10 microM), a potent inhibitor of calcineurin activity, was tested in brain slices and found not to alter tau mobility. However, combinations of FK-520 (5 microM) and okadaic acid (100 nM) caused tau mobility shifts similar to those seen after 10 microM okadaic acid treatment; similar results were seen using the calcineurin-selective inhibitor cypermethrin. Treatment of human slices with 10 microM okadaic acid decreased both protein phosphatase 2A and calcineurin activity; FK-520 inhibited only protein phosphatase 2B activity. A proposed tau-directed kinase, 42-kDa mitogen-activated protein kinase (p42mapk), was activated by okadaic acid (> 100 nM) but not FK-520 (5 microM). Nerve growth factor (100 ng/ml) activated p42mapk, particularly when used in combination with 100 nM okadaic acid; changes in tau mobility were seen when this kinase was activated. Forskolin (2 microM) antagonized the effects of nerve growth factor on both p42mapk activity and tau phosphorylation; forskolin alone had little effect on PHF-like tau formation induced by phosphatase inhibitors. These results outline complex interactions between tau-directed protein kinases and protein phosphatases and suggest potential sites for therapeutic intervention.


Subject(s)
Brain/metabolism , Phosphoprotein Phosphatases/physiology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , tau Proteins/metabolism , Amino Acid Sequence , Calcineurin , Calmodulin-Binding Proteins/analysis , Colforsin/pharmacology , Humans , Mitogen-Activated Protein Kinase 1 , Molecular Sequence Data , Nerve Growth Factors/pharmacology , Phosphoprotein Phosphatases/analysis , Phosphorylation , Protein Phosphatase 2
SELECTION OF CITATIONS
SEARCH DETAIL
...