Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
Acta Cir Bras ; 33(8): 703-712, 2018 Aug.
Article En | MEDLINE | ID: mdl-30208132

PURPOSE: To assess the action of vitamin C on the expression of 84 oxidative stress related-genes in cultured skin fibroblasts from burn patients. METHODS: Skin samples were obtained from ten burn patients. Human primary fibroblasts were isolated and cultured to be distributed into 2 groups: TF (n = 10, fibroblasts treated with vitamin C) and UF (n = 10, untreated fibroblasts). Gene expression analysis using quantitative polymerase chain reaction array was performed for comparisons between groups. RESULTS: The comparison revealed 10 upregulated genes as follows: arachidonate 12-lipoxygenase (ALOX12), 24-dehydrocholesterol reductase (DHCR24), dual oxidase 1 (DUOX1), glutathione peroxidase 2 (GPX2), glutathione peroxidase 5 (GPX5), microsomal glutathione S-transferase 3 (MGST3), peroxiredoxin 4 (PRDX4), phosphatidylinositol-3,4,5-trisphosphate dependent Rac exchange factor 1 (P-REX1), prostaglandin-endoperoxide synthase 1 (PTGS1), and ring finger protein 7 (RNF7). CONCLUSION: Cultured fibroblasts obtained from burn patients and treated with vitamin C resulted in 10 differentially expressed genes, all overexpressed, with DUOX1, GPX5, GPX2 and PTGS1 being of most interest.


Ascorbic Acid/pharmacology , Burns/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression/drug effects , Oxidative Stress/drug effects , Adult , Arachidonate 12-Lipoxygenase/analysis , Arachidonate 12-Lipoxygenase/drug effects , Burns/drug therapy , Cells, Cultured , Cross-Sectional Studies , Cyclooxygenase 1/analysis , Cyclooxygenase 1/drug effects , Dual Oxidases/analysis , Dual Oxidases/drug effects , Female , Glutathione Peroxidase/analysis , Glutathione Peroxidase/drug effects , Glutathione Transferase/analysis , Glutathione Transferase/drug effects , Guanine Nucleotide Exchange Factors/analysis , Guanine Nucleotide Exchange Factors/drug effects , Humans , Male , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/drug effects , Oxidoreductases Acting on CH-CH Group Donors/analysis , Oxidoreductases Acting on CH-CH Group Donors/drug effects , Peroxiredoxins/analysis , Peroxiredoxins/drug effects , Real-Time Polymerase Chain Reaction , Reference Values , Reproducibility of Results , Skin/drug effects , Skin/pathology , Statistics, Nonparametric , Ubiquitin-Protein Ligases/analysis , Ubiquitin-Protein Ligases/drug effects , Young Adult
2.
Acta cir. bras ; 33(8): 703-712, Aug. 2018. tab, graf
Article En | LILACS | ID: biblio-949375

Abstract Purpose: To assess the action of vitamin C on the expression of 84 oxidative stress related-genes in cultured skin fibroblasts from burn patients. Methods: Skin samples were obtained from ten burn patients. Human primary fibroblasts were isolated and cultured to be distributed into 2 groups: TF (n = 10, fibroblasts treated with vitamin C) and UF (n = 10, untreated fibroblasts). Gene expression analysis using quantitative polymerase chain reaction array was performed for comparisons between groups. Results: The comparison revealed 10 upregulated genes as follows: arachidonate 12-lipoxygenase (ALOX12), 24-dehydrocholesterol reductase (DHCR24), dual oxidase 1 (DUOX1), glutathione peroxidase 2 (GPX2), glutathione peroxidase 5 (GPX5), microsomal glutathione S-transferase 3 (MGST3), peroxiredoxin 4 (PRDX4), phosphatidylinositol-3,4,5-trisphosphate dependent Rac exchange factor 1 (P-REX1), prostaglandin-endoperoxide synthase 1 (PTGS1), and ring finger protein 7 (RNF7). Conclusion: Cultured fibroblasts obtained from burn patients and treated with vitamin C resulted in 10 differentially expressed genes, all overexpressed, with DUOX1, GPX5, GPX2 and PTGS1 being of most interest.


Humans , Male , Female , Adult , Young Adult , Ascorbic Acid/pharmacology , Burns/pathology , Gene Expression/drug effects , Oxidative Stress/drug effects , Fibroblasts/drug effects , Fibroblasts/pathology , Reference Values , Skin/pathology , Arachidonate 12-Lipoxygenase/analysis , Arachidonate 12-Lipoxygenase/drug effects , Burns/drug therapy , Cells, Cultured , Cross-Sectional Studies , Statistics, Nonparametric , Ubiquitin-Protein Ligases/analysis , Oxidoreductases Acting on CH-CH Group Donors/analysis , Cyclooxygenase 1/analysis , Cyclooxygenase 1/drug effects , Peroxiredoxins/analysis , Real-Time Polymerase Chain Reaction , Dual Oxidases/analysis , Dual Oxidases/drug effects , Glutathione Peroxidase/analysis , Glutathione Peroxidase/drug effects
3.
Cell Physiol Biochem ; 44(5): 1949-1964, 2017.
Article En | MEDLINE | ID: mdl-29224009

BACKGROUND/AIMS: This study was conducted to investigate the relationship between differentially expressed proteins (DEPs) and the pathogenesis of oleic acid (OA)-induced acute lung injury (ALI) in mice. METHODS: Eight-week-old male C57BL/6 mice were injected with OA through the tail vein and sacrificed 6 hours after OA administration to identify protein expression levels in lung tissue using isobaric tags for relative and absolute quantification (iTRAQ) technology. Then, DEPs such as antithrombin III (AT III), 12-lipoxygenase (12-LO), dedicator of cytokinesis 2 (DOCK2), polycystin-2 and plasminogen were identified by western blotting. Subsequently, we focused on investigating the effect of AT III on endothelial integrity using siRNA interference technology. The levels of IL-6, IL-1ß, TNF-α and TGF-ß expression were detected using an enzyme-linked immunosorbent assay (ELISA). Alterations in the tight junction component ZO-1 and the phosphorylation of myosin light chain (pMLC) were determined by western blotting. The stress fiber F-actin were also detected by immunofluorescence staining. In addition, endothelial permeability was determined via a transwell permeability assay. RESULTS: A total of 5152 proteins were found to be expressed in lung tissues from the OA-treated and saline-treated mice. Among these proteins, 849 were differentially expressed between the two groups, including 545 upregulated and 304 downregulated proteins. After AT III knockdown, the levels of inflammatory factors and endothelial permeability were elevated, the expression of ZO-1 was decreased, and the expression of F-actin and pMLC was increased. All these results illustrated that AT III knockdown exaggerated the disruption of endothelial integrity mediated by OA. CONCLUSION: These findings using iTRAQ technology demonstrate, for the first time, differences in the lung tissue expression levels of proteins between OA-treated mice and saline-treated mice. This study reveals that 12-LO, DOCK2 and especially AT III may be candidate biomarkers for OA-induced acute lung injury.


Acute Lung Injury/chemically induced , Down-Regulation/drug effects , Oleic Acid/toxicity , Proteomics , Up-Regulation/drug effects , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Antithrombin III/antagonists & inhibitors , Antithrombin III/genetics , Antithrombin III/metabolism , Arachidonate 12-Lipoxygenase/analysis , Arachidonate 12-Lipoxygenase/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Cell Line , GTPase-Activating Proteins/analysis , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , RNA Interference , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
J Dent Res ; 93(3): 300-5, 2014 Mar.
Article En | MEDLINE | ID: mdl-24389810

Resolvins are potent anti-inflammatory mediators derived from ω-3 fatty acids. Results from our previous studies indicated that resolvin D1 (RvD1) blocks pro-inflammatory responses in salivary glands. Furthermore, RvD1 enhances salivary epithelial integrity, demonstrating its potential use for the restoration of salivary gland function in Sjögren's syndrome (SS). We investigated whether the RvD1 biosynthetic machinery (e.g., cytosolic phospholipase A2, calcium-independent phospholipase A2, 12/15 and 5-lipoxygenase) is expressed in mouse submandibular glands (mSMG), using qPCR and Western blot analyses. Additionally, we determined the localization of RvD1 biosynthetic machinery in mSMG and human minor salivary glands (hMSG), with and without SS, using confocal microscopy. Finally, we measured RvD1 levels in cell supernatants from mSMG cell cultures and freshly isolated mSMG cells, with and without SS, using ELISA. Our results indicate that: (1) RvD1 machinery is expressed in mouse and human salivary glands; (2) polar distribution of RvD1 biosynthetic machinery is lost in hMSG with SS; (3) RvD1 levels in mSMG cell culture supernatants increased with time; and (4) RvD1 levels in mSMG cell supernatants, with and without SS, were similar. These studies demonstrate that the RvD1 biosynthesis machinery is expressed and functional in salivary glands with and without SS.


Docosahexaenoic Acids/biosynthesis , Submandibular Gland/metabolism , Animals , Arachidonate 12-Lipoxygenase/analysis , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/analysis , Arachidonate 15-Lipoxygenase/metabolism , Arachidonate 5-Lipoxygenase/analysis , Arachidonate 5-Lipoxygenase/metabolism , Cell Culture Techniques , Cell Membrane/enzymology , Cells, Cultured , Cytosol/enzymology , Docosahexaenoic Acids/analysis , Epithelium/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Inbred Strains , Phospholipases A2, Calcium-Independent/analysis , Phospholipases A2, Calcium-Independent/metabolism , Phospholipases A2, Cytosolic/analysis , Phospholipases A2, Cytosolic/metabolism , Salivary Glands, Minor/metabolism , Salivary Glands, Minor/pathology , Sjogren's Syndrome/metabolism , Sjogren's Syndrome/pathology , Submandibular Gland/cytology
5.
Dev Neurosci ; 35(2-3): 140-54, 2013.
Article En | MEDLINE | ID: mdl-23838566

Oxidative stress involving premyelinating oligodendrocytes (OLs) is a major factor in the pathogenesis of preterm white matter injury. In animal and cell culture studies, activation of the lipid-oxidizing enzyme 12/15-lipoxygenase (12/15-LOX) plays a central role as an inflammatory mediator in the pathology of oxidative stress and OL cell death, as well as ischemia and neuronal death. The role of 12/15-LOX, however, is unclear in the developing human brain. The mechanism of 12/15-LOX involves the production of reactive oxygen species through the metabolism of arachidonic acid, as well as direct detrimental effects on organelle membranes. Here we tested the hypothesis that the density of 12/15-LOX-expressing cells is increased in periventricular leukomalacia (PVL). Using immunocytochemistry (ICC) in human paraffin-embedded tissue, 12/15-LOX expression was seen in macrophages of the focally necrotic lesions in the periventricular white matter, as well as in glial cells throughout the surrounding white matter with reactive gliosis. Interestingly, no significant 12/15-LOX expression was detected in neurons in the cerebral cortex overlying the damaged white matter. Using a scoring system from 0 to 3, we assessed the density of 12/15-LOX-expressing cells in diffusely gliotic white matter from 20 to 43 postconceptional (PC) weeks in 19 PVL cases (median = 36 PC weeks) and 10 control (non-PVL) cases (median = 34 PC weeks). The density of 12/15-LOX-positive cells was significantly increased in the diffuse component of PVL (score = 1.17 ± 0.15) compared to controls (score = 0.48 ± 0.21; p = 0.014). Using double-label ICC, 12/15-LOX was observed in PVL in OLs of the O4 and O1 premyelinating stages, as well as in mature OLs as determined with the mature OL marker adenomatous polyposis coli (APC). In addition, 12/15-LOX expression was present in a population of CD68-positive activated microglia. There was no 12/15-LOX expression in reactive astrocytes. Finally we observed terminal deoxynucleotide transferase dUTP nick end-labeling-positive cells within the white matter of PVL that expressed 12/15-LOX and/or within close proximity of 12/15-LOX-positive cells. Our data support a role for 12/15-LOX activation as an inflammatory mediator of injury in PVL, with a contribution of 12/15-LOX to PVL-induced damage to or cell death of OLs, including those at the O1 and O4 stages.


Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Leukomalacia, Periventricular/enzymology , Microglia/enzymology , Oligodendroglia/enzymology , Arachidonate 12-Lipoxygenase/analysis , Arachidonate 15-Lipoxygenase/analysis , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Infant, Newborn , Leukomalacia, Periventricular/pathology
6.
Arch Otolaryngol Head Neck Surg ; 134(8): 825-31, 2008 Aug.
Article En | MEDLINE | ID: mdl-18711055

OBJECTIVE: To evaluate the expression of cyclooxygenase (COX) and lipoxygenase (LO) enzymes in the sinonasal mucosa of patients with cystic fibrosis (CF). DESIGN: Immunohistochemical staining of archived tissue. PARTICIPANTS: Specimens from 9 patients with CF were analyzed; control specimens were obtained from 4 patients without a history of CF or rhinosinusitis. INTERVENTIONS: Expression of the enzymes COX-1, COX-2, 5-LO, 12-LO, and 15-LO was evaluated with the use of immunohistochemical techniques in archived sinonasal mucosal tissue from patients with CF. These results were compared with those of the control group. RESULTS: We noted the characteristic staining patterns of epithelium and submucosal glands for each enzyme. Statistically significant (P < .05) differences between control and CF specimens were noted in the staining intensity of columnar epithelium for COX-2 (cytoplasm) and 12-LO (cytoplasm and nucleus) and of submucosal glands for COX-2 (cytoplasm) and 12-LO (cytoplasm). No significant differences were noted for the staining intensity of COX-1, 5-LO, or 15-LO between the groups. CONCLUSIONS: Significant differences in sinonasal mucosal expression of COX-2 and 12-LO enzymes exist between patients with CF and controls. This suggests a difference in arachidonic acid metabolism between these 2 groups.


Cystic Fibrosis/pathology , Lipoxygenase/analysis , Prostaglandin-Endoperoxide Synthases/analysis , Rhinitis/pathology , Sinusitis/pathology , Adolescent , Adult , Arachidonate 12-Lipoxygenase/analysis , Arachidonate 15-Lipoxygenase/analysis , Arachidonate 5-Lipoxygenase/analysis , Child , Child, Preschool , Cyclooxygenase 1/analysis , Cyclooxygenase 2/analysis , Female , Humans , Male , Middle Aged , Nasal Mucosa/pathology , Nasal Polyps/pathology , Reference Values
8.
Anal Biochem ; 322(2): 198-207, 2003 Nov 15.
Article En | MEDLINE | ID: mdl-14596828

A method using confocal Raman microspectroscopy for the detection of cellular proteins in single intact cells was developed. Two approaches were used to improve the detection of these cellular components. First, compounds with high Raman scattering were investigated for potential use as Raman labels. Raman labels were conjugated to either biomolecules or biotin and used as markers in the detection of cellular enzymes and receptors. Second, silver colloids were used to increase the surface-enhanced Raman scatter (SERS) of these Raman labels. Cresyl violet and dimethylaminoazobenzene are Raman labels that provide very sensitive SERS detection by a confocal Raman microscope with a HeNe laser at wavelength of 632.8 nm. The detection of 12-lipoxygenase and cyclooxygenase-1 in single bovine coronary artery endothelial cells and the binding of angiotensin II to its receptors in zona glomerulosa cells was demonstrated.


Coloring Agents/chemistry , Proteins/analysis , Spectrum Analysis, Raman/methods , Angiotensin II/analysis , Animals , Arachidonate 12-Lipoxygenase/analysis , Benzoxazines , Biotin/chemistry , Cattle , Cells, Cultured , Citrates/chemistry , Colloids/chemistry , Coronary Vessels/cytology , Coronary Vessels/enzymology , Cyclooxygenase 1 , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Fluorescent Antibody Technique/methods , Isoenzymes/analysis , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Oxazines/chemistry , Prostaglandin-Endoperoxide Synthases/analysis , Proteins/chemistry , Receptors, Angiotensin/metabolism , Sensitivity and Specificity , Silver Compounds/chemistry , Zona Glomerulosa , p-Dimethylaminoazobenzene/chemistry
9.
Arch Biochem Biophys ; 386(2): 268-74, 2001 Feb 15.
Article En | MEDLINE | ID: mdl-11368351

Formation of the 12R-lipoxygenase product, 12R-hydroperoxyeicosatetraenoic acid (12R-HPETE), has been detected previously only in human skin (Boeglin et al. (1998) Proc. Natl. Acad. Sci. USA 95, 6744). The unexpected appearance of an EST sequence (AA649213) for human 12R-lipoxygenase from germinal center B lymphocytes purified from human tonsils prompted our search for the existence of the enzyme in this novel source. Incubation of [1-14C]arachidonic acid with homogenates of human tonsillar tissue yielded mixtures of radiolabeled 12-HETE and 15-HETE. Stereochemical analysis showed varying ratios of 12S- and 12R-HETE, while 15-HETE was exclusively of the S-configuration. Using stereospecifically labeled [10S-3H]- and [10R-3H]arachidonic acid substrates we detected pro-R hydrogen abstraction at carbon 10 associated with formation of 12R-HETE. This mechanistic evidence implicates a 12R-lipoxygenase in the biosynthesis of 12R-HETE. The mRNA for the enzyme was identified in tonsils by RT-PCR and Northern analysis. The cellular distribution was established by in situ hybridization. Unexpectedly, hybridization was not observed in the lymphocytes of the germinal centers. Specific reaction was restricted to squamous epithelial cells, including the epithelium lining the tonsillar crypts. In this location the 12R-lipoxygenase might help regulate differentiation of the epithelium or participate in lymphocyte- epithelial cell interactions.


Arachidonate 12-Lipoxygenase/metabolism , Palatine Tonsil/enzymology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Arachidonate 12-Lipoxygenase/analysis , Arachidonate 12-Lipoxygenase/genetics , Child , Child, Preschool , Chromatography, High Pressure Liquid , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Expressed Sequence Tags , Germinal Center/enzymology , Humans , Hydroxyeicosatetraenoic Acids/metabolism , In Situ Hybridization , Palatine Tonsil/cytology , Palatine Tonsil/metabolism , Prostaglandins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Comp Biochem Physiol B Biochem Mol Biol ; 127(4): 563-73, 2000 Dec.
Article En | MEDLINE | ID: mdl-11281273

12-Lipoxygenase (12-LO) in bullfrog (Rana catesbeiana) erythrocytes was purified partially by ion exchange chromatography and affinity chromatography. Bullfrog 12-LO was a single chain protein with a pI of 7.1-7.8 and MW of 7.77 kDa. This enzyme did not show typical Michaelis Menten type kinetics. At low substrate concentrations, it had a lag phase and at higher substrate concentrations, the activity was inhibited. The product of linoleic acid (LA), 13-hydroperoxy-9, 11-octadecadienoic acid (13-HpODE), was an activator for the enzyme. When arachidonic acid (AA) was used as substrate, 13-HpODE also affected the Km of bullfrog 12-LO towards AA. The affinity of LA towards bullfrog 12-LO was higher than the affinity of AA. Suicide inactivation was much more rapid than that of any mammalian 12-LO reported. Hemoglobin (Hb) inhibited the activity of 12-LO partially and removing Hb eliminated this inhibition. Both Hb and Met-Hb inhibited the 12-LO activity but did not denatured completely the Hb, suggesting that the inhibition was a direct interaction between 12-LO and Hb protein chain and was not due to competition between 12-LO and Hb for oxygen. This study characterizes bullfrog 12-LO with respect to stability, optimal pH, suicide inactivation and interaction with Hb and provides important evolutionary information about this enzyme.


Arachidonate 12-Lipoxygenase/analysis , Arachidonate 12-Lipoxygenase/isolation & purification , Arachidonic Acid/metabolism , Erythrocytes/enzymology , Rana catesbeiana/blood , Animals , Chromatography, Affinity , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Chromatography, Thin Layer , Hemoglobins/metabolism , Hydrogen-Ion Concentration , Isoelectric Focusing , Linoleic Acids/metabolism , Lipid Peroxides/metabolism , Molecular Weight , Subcellular Fractions/metabolism , Substrate Specificity , Temperature
11.
Cell Tissue Res ; 288(2): 327-34, 1997 May.
Article En | MEDLINE | ID: mdl-9082968

Arachidonate 12-lipoxygenase enzyme oxygenates the position 12 of arachidonic acid and produces 12-hydroperoxy-arachidonic acid. Mouse keratinocytes were transiently transfected with an expression vector of human platelet 12-lipoxygenase cDNA. The cells were homogenized, and the subcellular localization of the enzyme was examined by differential centrifugation. The 12-lipoxygenase activity was detected predominantly in the particulate fractions. In contrast, immunoelectron microscopy detected the enzyme mainly in the cytoplasm of the transfected cell, but not in the nucleus, subcellular organelles or plasma membrane. To explain the discrepancy between these findings, we performed an electron-microscopic examination of the 176000 g pellet of the keratinocyte homogenate. The pellet contained mainly insoluble proteins such as keratin but not membrane structures such as the plasma membrane. Thus, it is possible that the enzyme was localized originally in the cytoplasm of the keratinocyte, and found in the particulate fractions due to its association with insoluble proteins during fractionation procedures. Unique structural changes were observed in the transfected keratinocytes. The nucleus had very scant karyoplasm and coarse fibrillary structures. When the keratinocytes were transfected with a mutant 12-lipoxygenase cDNA or a vector without 12-lipoxygenase cDNA, these structural changes were not observed.


Arachidonate 12-Lipoxygenase/analysis , Gene Expression Regulation, Enzymologic/physiology , Keratinocytes/enzymology , Subcellular Fractions/enzymology , Transfection , Animals , Arachidonate 12-Lipoxygenase/genetics , Blood Platelets/enzymology , Genetic Vectors , Humans , Immunohistochemistry , Keratinocytes/ultrastructure , Mice , Microscopy, Electron
12.
Am J Hypertens ; 10(4 Pt 1): 371-8, 1997 Apr.
Article En | MEDLINE | ID: mdl-9128202

We have previously demonstrated that administration of inhibitors of the lipoxygenase (LO) pathway of arachidonic acid metabolism lowers blood pressure in hypertensive rats. In addition, we have shown that LO inhibition attenuates pressor agonist-induced vascular reactivity in vitro and calcium mobilization in cultured vascular smooth muscle cells (VSMC). To further elucidate the relationship between elevated LO activity and hypertension, 4, 8, and 12 week old hypertensive SHR were compared with age-matched Wistar-Kyoto (WKY) rats for plasma 12(S)-hydroxyeicosatetraenoic acid (12-HETE) concentration. 12-HETE levels were significantly elevated in the SHR compared to the WKY (SHR elevated by 154%, 159%, and 272% compared to WKY at 4, 8, and 12 weeks, respectively, P < .01 for all ages). There were no differences in plasma potassium levels between SHR and WKY at any of the ages tested. Plasma aldosterone levels and plasma renin activity were in the normal range at the three ages. At 12 weeks of age, both serum (4.72 +/- 0.23 v 2.18 +/- 0.33 microg/mL, P < .01), and aortic smooth muscle 12-HETE levels (0.94 +/- 0.09 v 0.66 +/- 0.08 microg/mg protein, P < .05) were elevated in SHR compared with WKY. The 12 week old SHR were given a bolus of the LO inhibitor 5,8,11-eicosatriynoic acid (ETI, 7 mg/kg, intravenously) and blood pressure measured after 20 min. ETI reduced mean systolic blood pressure from 175.8 +/- 4.2 to 141.6 +/- 5.9 mm Hg (P < .05). To investigate these effects of HETEs, cultured vascular smooth muscle cells were pretreated for 1 min with 12(S)HETE and then challenged with angiotensin II (AngII). The addition of 12(S)HETE increased AngII-induced intracellular calcium levels in normal cultured rat vascular smooth muscle cells by 78% compared to vehicle (P < .05). Thus, LO products, which are high in SHR, may contribute to vascular tone through alterations in the intracellular calcium signal by potentiating calcium responses to pressors such as Ang II.


Arachidonate 12-Lipoxygenase/analysis , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/blood , Aging , Animals , Calcium/metabolism , Cells, Cultured , Hypertension/physiopathology , Male , Muscle, Smooth, Vascular/physiopathology , Rats , Rats, Inbred SHR , Rats, Inbred WKY
14.
Exp Cell Res ; 228(2): 197-205, 1996 Nov 01.
Article En | MEDLINE | ID: mdl-8912711

Human A431 epidermoid carcinoma cells express 12-lipoxygenase enzymatic activity. However, the isoform identity based on cDNA sequence data is not known. Further, the simultaneous characterization of the intracellular distribution of 12-lipoxygenase protein and activity is lacking. Here we report that the cDNA sequence from RT-PCR-amplified 12-lipoxygenase mRNA is identical with the platelet-type 12-lipoxygenase isoform, and the leukocyte-type isoform of 12-lipoxygenase is not expressed in A431 cells. The predominant amount (78%) of 12-lipoxygenase protein resides in the cytosol. In contrast, the predominant (98%) 12-lipoxygenase activity is localized in the membrane fraction. Western blot and immunofluorescence data demonstrate that epidermal growth factor increases total cellular 12-lipoxygenase protein and enhances the association of 12-lipoxygenase protein with perinuclear or nuclear membrane sites. In addition, epidermal growth factor stimulates 12-lipoxygenase activity resulting in generation of 12(S)-hydroxyeicosatetraenoic acid from cellular arachidonate. In contrast, both 12-lipoxygenase protein and activity decrease approximately 80% within 24 h during serum starvation. The recovery of 12-lipoxygenase expression in serum-deprived cells can be induced by readdition of epidermal growth factor or serum. Further, the basal expression of 12-lipoxygenase depends on signal pathways requiring protein tyrosine kinase activity, since genistein, herbimycin A, and tyrphostin 25 reduce the expression of 12-lipoxygenase protein in A431 cells.


Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 12-Lipoxygenase/genetics , Gene Expression Regulation, Enzymologic , Arachidonate 12-Lipoxygenase/analysis , Base Sequence , Blood Platelets/enzymology , Carcinoma, Squamous Cell , Cell Line , Cell Membrane/enzymology , Culture Media, Serum-Free , Cytosol/enzymology , DNA Primers , DNA, Complementary , Epidermal Growth Factor/pharmacology , Exons , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/biosynthesis , Sequence Homology, Nucleic Acid , Signal Transduction , Transcription, Genetic
15.
J Endocrinol ; 148(1): 33-41, 1996 Jan.
Article En | MEDLINE | ID: mdl-8568469

Arachidonate 12-lipoxygenase, which oxygenates positions 12 and 13 of arachidonic and linoleic acids, is present in porcine anterior pituitary cells. Colocalization of the 12-lipoxygenase with various pituitary hormones was examined by immunohistochemical double-staining using antibodies against 12-lipoxygenase and various anterior pituitary hormones. Under light microscopy, approximately 7% of the cells producing luteinizing hormone (LH) and follicle-stimulating hormone (FSH) were positive for 12-lipoxygenase, whereas the enzyme was detected in less than 2% of the cells producing thyrotrophin, prolactin, growth hormone (GH), and adrenocorticotrophin. In an attempt to examine the participation of 12-lipoxygenase metabolites in pituitary hormone release, we incubated the primary culture of porcine anterior pituitary cells with 12-hydroperoxy-arachidonic acid or 13-hydroperoxy-linoleic acid. Significant stimulation of LH and FSH release by these hydroperoxides was observed at 10 microM in a time-dependent manner. At doses around 10 microM these compounds produced responses of similar magnitude to 1 nM gonadotrophin-releasing hormone (GnRH), but higher concentrations (30 microM) of the compounds were required for GH release. In contrast, 12-hydroxy-arachidonic and 13-hydroxy-linoleic acids were almost ineffective. Furthermore, the gonadotrophin release by 1 nM GnRH was inhibited by nordihydroguaiaretic acid (a lipoxygenase inhibitor) with an IC50 of about 5 microM. Thus, the hydroperoxy (but not hydroxy) products of 12-lipoxygenase may be involved in the release of pituitary hormones especially LH and FSH.


Arachidonate 12-Lipoxygenase/analysis , Lipid Peroxides , Pituitary Gland, Anterior/enzymology , Swine/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid , Animals , Antithrombins/pharmacology , Arachidonate 12-Lipoxygenase/metabolism , Cells, Cultured , Chromatography, High Pressure Liquid , Fatty Acids, Unsaturated/analysis , Follicle Stimulating Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Growth Hormone/metabolism , Hydroxyeicosatetraenoic Acids/pharmacology , Immunohistochemistry , Leukotrienes/pharmacology , Linoleic Acids/pharmacology , Luteinizing Hormone/metabolism , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Time Factors
16.
J Histochem Cytochem ; 43(3): 237-44, 1995 Mar.
Article En | MEDLINE | ID: mdl-7868854

Arachidonate 12-lipoxygenase is an enzyme that oxygenates the 12 position of arachidonic acid to produce its 12-hydroperoxy derivative. We were interested in the tissue distribution and subcellular localization of the platelet-type 12-lipoxygenase, which is distinguished from the leukocyte type by several criteria. Antiserum was raised in rabbits against purified recombinant arachidonate 12-lipoxygenase of human platelets. When mouse bone marrow cells and lung were immunostained and observed by light and electron microscopy, the positively stained cells were platelets, megakaryocytes, and eosinophils. 12-Lipoxygenase was localized in the cytoplasm of platelets but was hardly detectable in the plasma membrane and intracellular organelles. The enzyme was found in the cytoplasm of immature megakaryocytes with kidney-shaped nuclei and a few profiles of demarcation membranes, as well as in the mature form with well-developed demarcation membranes. These results indicated the expression of 12-lipoxygenase at an early stage in the course of megakaryocytopoiesis.


Arachidonate 12-Lipoxygenase/blood , Blood Platelets/enzymology , Eosinophils/enzymology , Hematopoietic Stem Cells/enzymology , Megakaryocytes/enzymology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid , Animals , Antibodies , Arachidonate 12-Lipoxygenase/analysis , Blood Platelets/ultrastructure , Bone Marrow Cells , Chromatography, High Pressure Liquid , Eosinophils/ultrastructure , Hematopoietic Stem Cells/ultrastructure , Humans , Hydroxyeicosatetraenoic Acids/analysis , Hydroxyeicosatetraenoic Acids/blood , Immunohistochemistry/methods , Linoleic Acids/analysis , Linoleic Acids/blood , Lung/cytology , Lung/enzymology , Megakaryocytes/ultrastructure , Mice , Microscopy, Immunoelectron/methods , Recombinant Proteins/analysis
17.
Prostaglandins ; 49(1): 49-62, 1995 Jan.
Article En | MEDLINE | ID: mdl-7792391

Recently we demonstrated that Lewis lung (3LL) tumor cells express 12-lipoxygenase (12-LOX) mRNA and protein, respectively. In this study we partially sequenced the 12-LOX cDNA after reverse-transcription polymerase chain reaction amplification of 12-LOX mRNA from cultured 3LL cells. Comparison with platelet and leukocyte 12-LOX indicates that 3LL 12-LOX is identical with the platelet-type enzyme at least within the sequenced region. Further, we investigated the intracellular distribution of both 12-LOX enzyme protein and its activity which are prerequisites for understanding 12-LOX regulation. 12-LOX activity was monitored via the production of 12-hyroxyeicosatetraenoic acid from 3LL cells and their subcellular fractions using reverse-phase high performance liquid chromatography. 12-LOX protein was measured by direct slot blot and by Western Blotting. In 3LL cells, both 12-LOX activity and 12-LOX protein were predominantly localized in the cytosol. This 12-LOX activity was optimal at 37 degrees C. However at 24 degrees C and 10 degrees C, it showed 87% and 61% of this activity, respectively, thus differing distinctly from 12-LOX in platelets or rat basophilic leukemia cells. Incubation of 3LL cell homogenates with 0-100 microM free Ca2+ and subsequent separate analyses of cytosol and membrane fractions indicated that, as in platelets, an increase in intracellular free Ca2+ caused a loss of cytosolic 12-LOX activity. However, no significant Ca(2+)-induced increase in membrane-associated 12-LOX activity was observed under these conditions in 3LL cells. In contrast, at the 12-LOX protein level we observed a Ca(2+)-dependent loss in the cytosol and a concomitant increase in the membrane fraction. Thus, we suggest that 12-LOX in 3LL cells undergoes rapid translocation from cytosol to membrane in a Ca(2+)-dependent manner, but is no longer active or becomes inactivated at the membrane site.


Arachidonate 12-Lipoxygenase/analysis , Calcium/pharmacology , Carcinoma, Lewis Lung/enzymology , Carcinoma, Lewis Lung/pathology , Cell Membrane/enzymology , Cytosol/enzymology , Amino Acid Sequence , Animals , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 12-Lipoxygenase/genetics , Base Sequence , Biological Transport , Blood Platelets/enzymology , Calcium/metabolism , Carcinoma, Lewis Lung/genetics , Cell Membrane/metabolism , Cytosol/metabolism , DNA, Complementary , Enzyme Activation , Leukocytes/enzymology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/analysis , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Temperature , Tumor Cells, Cultured/enzymology
18.
Cancer Res ; 54(6): 1574-9, 1994 Mar 15.
Article En | MEDLINE | ID: mdl-7511046

12(S)-Hydroxyeicosatetraenoic acid [12(S)-HETE] is the 12-lipoxygenase metabolite of arachidonic acid. Previously, we have demonstrated that exogenous 12(S)-HETE can activate protein kinase C, increase cell surface expression of integrins, enhance adhesion, induce endothelial cell retraction, and increase experimental metastasis of tumor cells. Because of these prominent effects of exogenous 12(S)-HETE on tumor cell metastatic potential, it is important to determine whether there is endogenous 12(S)-HETE production by tumor cells. In the present study, mRNAs from human, rat, and mouse platelets as well as human colon carcinoma (Clone A), rat Walker carcinoma (W256), and mouse melanoma (B16a) and lung carcinoma (3LL) were reverse transcribed and amplified by polymerase chain reaction with platelet 12-lipoxygenase specific primers. Identity of the polymerase chain reaction fragments was confirmed by sequencing. 12-Lipoxygenase protein was detected by Western blotting. Tumor cell-derived 12-HETE was determined by reverse phase-high performance liquid chromatography analysis. In addition, the effect of endogenous 12(S)-HETE on tumor cells was studied by using a platelet-type 12-lipoxygenase selective inhibitor (N-benzyl-N-hydroxy-5-phenylpentanamide). Our results suggest that some tumor cells express platelet-type 12-lipoxygenase mRNA, protein and metabolize arachidonic acid to 12(S)-HETE and that endogenous 12(S)-HETE, like the exogenous 12(S)-HETE, may play an important role in tumor cell adhesion to matrix in vitro and lung colonization in vivo.


Arachidonate 12-Lipoxygenase/analysis , Hydroxyeicosatetraenoic Acids/biosynthesis , Hydroxyeicosatetraenoic Acids/physiology , Neoplasm Metastasis/physiopathology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid , Amino Acid Sequence , Animals , Arachidonate 12-Lipoxygenase/physiology , Base Sequence , Blotting, Western , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Adhesion/physiology , Chromatography, High Pressure Liquid/methods , Extracellular Matrix/physiology , Humans , Lipoxygenase Inhibitors , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neoplasms, Experimental/enzymology , Polymerase Chain Reaction/methods , RNA-Directed DNA Polymerase/metabolism , Rats
19.
Biochim Biophys Acta ; 1210(3): 288-96, 1994 Jan 20.
Article En | MEDLINE | ID: mdl-8305483

Lipoxygenases of bovine and human corneal epithelia were investigated. The bovine epithelium contained an arachidonate 12-lipoxygenase and a 15-lipoxygenase. The 12-lipoxygenase was found in the microsomal fraction, while the 15-lipoxygenase was mainly present in the cytosol (100,000 x g supernatant). 12S-Hydroxyeicosatetraenoic acid (12S-HETE) and 15S-hydroxyeicosatetraenoic acid (15S-HETE) were identified by GC-MS and chiral HPLC. BW A4C, an acetohydroxamic acid lipoxygenase inhibitor, reduced the biosynthesis of 12S-HETE and 15S-HETE by over 90% at 10 microM. IC50 for the 12-lipoxygenase was 0.3 microM. The bovine corneal 12-lipoxygenase was compared with the 12-lipoxygenases of bovine platelets and leukocytes. All three enzymes metabolized 14C-labelled linoleic acid and alpha-linolenic acid poorly (5-16%) in comparison with [14C]arachidonic acid. [14C]Docosahexaenoic acid and [14C]4,7,10,13,16-docosapentaenoic acid appeared to be less efficiently converted by the corneal enzyme than by the platelet and leukocyte enzymes. Immunohistochemical analysis of the bovine corneal epithelium using a polyclonal antibody against porcine leukocyte 12-lipoxygenase gave positive staining. The cytosol of human corneal epithelium converted [14C]arachidonic acid to one prominent metabolite. The product co-chromatographed with 15S-HETE on reverse phase HPLC, straight phase HPLC and chiral HPLC. Our results suggest that human corneal epithelium contains a 15-lipoxygenase and that bovine corneal epithelium contains both a 15-lipoxygenase and a 12-lipoxygenase. The corneal 12-lipoxygenase appears to differ catalytically from earlier described bovine 12-lipoxygenases.


Arachidonate 12-Lipoxygenase/analysis , Arachidonate 15-Lipoxygenase/analysis , Cornea/enzymology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid , Animals , Arachidonic Acid/metabolism , Cattle , Chromatography, High Pressure Liquid , Epithelium/enzymology , Humans , Hydroxyeicosatetraenoic Acids/biosynthesis , Immunohistochemistry , Subcellular Fractions/enzymology
20.
J Lipid Res ; 34(9): 1505-14, 1993 Sep.
Article En | MEDLINE | ID: mdl-8228634

The extent to which fatty acid oxygenases are activated in the normal epidermis is not known. Characterization of the regio- and stereospecificity of the monohydroxylated derivatives of arachidonic and linoleic acid produced by human hair roots is needed to define the enzymatic origin of these compounds and to define a possible role for fatty acid oxygenases in growth, differentiation, and pathology of human hair. Hair roots epilated from normal human volunteers were incubated with radiolabeled arachidonic acid or linoleic acid and the monohydroxylated derivatives produced in vitro were characterized. Incubation of hair roots with 14C]arachidonic acid resulted in the production of 15(S)-[14C]hydroxyeicosatetraenoic acid and 12(S,R)-[14C]hydroxyeicosatetraenoic acid (mean S/R ratio, 2.5). 13(S)-[14C]hydroxyoctadecadienoic acid was the principal product of incubations with [14C]linoleic acid. No radiolabeled products were derived from incubations with heat-denatured hair roots. The fatty acid oxygenase activity of anagen hair roots was inhibited by nordihydroguaiaretic acid and was greatest in the hair root bulb. The strict S-stereospecificity and the regiospecificity of the n-6 oxygenase are strong evidence for the presence of a 15-lipoxygenase in human hair roots, similar to that identified in cultured human keratinocytes. The stereospecificity of the 12-HETE produced by human hair roots is not compatible with the sole action of 12-lipoxygenase.


Arachidonate 12-Lipoxygenase/analysis , Arachidonate 15-Lipoxygenase/analysis , Hair/enzymology , Chromatography, High Pressure Liquid , Female , Humans , Male
...