Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
2.
Acta Cir Bras ; 33(8): 703-712, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30208132

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.


Subject(s)
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
3.
Acta cir. bras ; 33(8): 703-712, Aug. 2018. tab, graf
Article in English | LILACS | ID: biblio-949375

ABSTRACT

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.


Subject(s)
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
4.
Brain Res ; 1678: 123-128, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29079502

ABSTRACT

12/15-Lipoxygenase (12/15-LOX) contributes to the brain damage after middle cerebral artery occlusion (MCAO) in the acute phase of stroke. The aim of this study was to investigate the effects of a 12/15-LOX inhibitor, LOXBlock-1(LB1), in mice using a FeCl3-induced permanent distal MCAO model and FeCl3-induced ischemia/thrombolysis with tPA. In order to induce permanent distal MCAO, 30% FeCl3 was used in C57BL6 mice. LB1 or DMSO treatments were applied intraperitoneally 2 h following MCAO. For FeCl3-induced ischemia/thrombolysis experiments, 10% FeCl3 was preferred so as to obtain reperfusion with tPA in CD1 mice. 4 h following ischemia either LB1 or DMSO and iv tPA was administered. Outcomes were NSS, weight loss, infarct volume, hemorrhage area and reperfusion rate. FeCl3-induced distal MCAO caused an increase in 12/15-LOX signal in the ischemic cortex with an increase in MDA2 and AIF immunoreactivity. LB1 treatment, applied 2 h after ischemia, significantly decreased the infarct volume at 24 h of permanent distal MCAO. Weight loss was also significantly reduced in LB1 treated group. Distal MCAO and tPA application with LB1 or DMSO showed that treatment significantly decreased the infarct volume and the hemorrhage area. The reperfusion rate in the LB1-treated group was surprisingly higher than in the DMSO group and NSS results were significantly improved. These data suggest that LB1 can be used as an adjuvant agent to tPA. This study not only shows the effects of LB1 treatment in distal MCAO but also confirms that FeCl3-induced MCAO model can be a useful tool to screen novel treatment options in stroke.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Brain Ischemia/drug therapy , Stroke/physiopathology , Animals , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/drug effects , Brain Ischemia/metabolism , Disease Models, Animal , Fibrinolytic Agents/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Lipoxygenase/drug effects , Lipoxygenase/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Reperfusion , Stroke/drug therapy , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/therapeutic use
5.
Ceska Slov Farm ; 63(1): 26-31, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24568335

ABSTRACT

Curcumin (diferuloylmethane) is an orange-yellow secondary metabolic compound from the rhizome of turmeric (Curcuma longa L.), a spice often found in curry powder. It is one of the major curcuminoids of turmeric. For centuries, curcumin has been used in some medicinal preparations or as a food colouring agent. A variety of enzymes that are closely associated with inflammation and cancer were found to be modulated by curcumin. This paper summarized the results of the inhibitory effect of curcumin and a Curcuma longa L. ethanolic extract on lipoxygenase from the rat lung cytosolic fraction. The positional specificity determination of arachidonic acid dioxygenation by RP- and SP-HPLC methods showed that in a purified enzyme preparation from the rat lung cytosol the specific form of lipoxygenase (LOX) is present exhibiting 12/15-LOX dual specificity (with predominant 15-LOX activity). The inhibitory activity of curcumin and Curcuma longa extract on LOX from cytosolic fraction of rat lung was expressed in the percentage of inhibition and as IC50. Lineweaver-Burk plot analysis has indicated that curcumin is the competitive inhibitor of 12/15 LOX from the rat lung cytosolic fraction.


Subject(s)
Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/drug effects , Curcuma/chemistry , Curcumin/pharmacology , Animals , Arachidonic Acid/chemistry , Curcumin/isolation & purification , Male , Rats , Rats, Wistar , Rhizome
6.
J Steroid Biochem Mol Biol ; 140: 1-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24269661

ABSTRACT

Primary cultures of human bone and vascular cells respond to vitamin D treatment by modulation of cell proliferation measured by DNA synthesis (DNA) and energy metabolism measured by creatine kinase specific activity (CK) via binding to vitamin D receptors (VDR) which are expressed in these cells. Vitamin D compounds also modulate the response to estradiol-17ß (E2) and the expression mRNAs of estrogen receptors (ERα and ERß), VDR, 25-hydroxy vitamin D3 1-α hydroxylase (1OHase) and lipoxygenases (12LO and 15LO). We now compared our newly synthesized analog: 1α,25-dihydroxy-9-methylene-19-norvitamin D3 JK152 (JK), on bone and vascular cells compared to other analogs. Human bone cell line SaOS2 respond to JK by increased DNA and stimulated CK dose-dependently, similar to the less-calcemic analogs CB 1093 (CB) and EB 1089 (EB). JK also up-regulated the response to E2 in terms of DNA and CK. JK inhibited DNA synthesis and increased CK in primary human vascular smooth muscle cells (VSMC) dose-dependently similar to EB and CB. JK up regulated the response to E2 in terms of CK with no effect on DNA. JK similar to CB and EB stimulated mRNA expression of VDR and ERα, 12LO and 15LO, with no effect on ERß and 1OHase mRNA expression in SaOS2 measured by real time PCR. Similar treatments of VSMC with JK, CB and EB stimulated 12LO and 15LO, VDR and ERα mRNA expression with no effect on ERß and 1OHase mRNA expression. The results presented here demonstrate that the new vitamin D less-calcemic analog JK is similar to other analogs in its effects on human cultured cells and therefore may be used in combined hormone replacement treatment (HRT) both in vitro and in vivo.


Subject(s)
Bone and Bones/drug effects , Calcitriol/analogs & derivatives , Muscle, Smooth, Vascular/drug effects , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/biosynthesis , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/drug effects , Bone and Bones/metabolism , Calcitriol/pharmacology , Cell Line , Cells, Cultured , Creatine Kinase/metabolism , DNA/biosynthesis , DNA/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/biosynthesis , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/metabolism , Receptors, Calcitriol/biosynthesis
7.
FASEB J ; 27(5): 1939-49, 2013 May.
Article in English | MEDLINE | ID: mdl-23382512

ABSTRACT

Previously, we observed significant increases in spinal 12-lipoxygenase (LOX) metabolites, in particular, hepoxilins, which contribute to peripheral inflammation-induced tactile allodynia. However, the enzymatic sources of hepoxilin synthase (HXS) activity in rats remain elusive. Therefore, we overexpressed each of the 6 rat 12/15-LOX enzymes in HEK-293T cells and measured by LC-MS/MS the formation of HXB3, 12-HETE, 8-HETE, and 15-HETE from arachidonic acid (AA) at baseline and in the presence of LOX inhibitors (NDGA, AA-861, CDC, baicalein, and PD146176) vs. vehicle-treated and mock-transfected controls. We detected the following primary intrinsic activities: 12-LOX (Alox12, Alox15), 15-LOX (Alox15b), and HXS (Alox12, Alox15). Similar to human and mouse orthologs, proteins encoded by rat Alox12b and Alox12e possessed minimal 12-LOX activity with AA as substrate, while eLOX3 (encoded by Aloxe3) exhibited HXS without 12-LOX activity when coexpressed with Alox12b or supplemented with 12-HpETE. CDC potently inhibited HXS and 12-LOX activity in vitro (relative IC50s: CDC, ~0.5 and 0.8 µM, respectively) and carrageenan-evoked tactile allodynia in vivo. Notably, peripheral inflammation significantly increased spinal eLOX3; intrathecal pretreatment with either siRNA targeting Aloxe3 or an eLOX3-selective antibody attenuated the associated allodynia. These findings implicate spinal eLOX3-mediated hepoxilin synthesis in inflammatory hyperesthesia and underscore the importance of developing more selective 12-LOX/HXS inhibitors.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Hyperalgesia/etiology , Intramolecular Oxidoreductases/metabolism , Animals , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/drug effects , HEK293 Cells , Humans , Lipoxygenase Inhibitors/pharmacology , Male , Rats
8.
Brain Res ; 1473: 227-35, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22867942

ABSTRACT

Hydroxysafflor yellow A (HSYA) is the main component of the safflower yellow pigments, the aqueous extract of safflower florets. We report here an experimental study for evaluating HSYA for their neuroprotective qualities on rats subjected to middle cerebral artery occlusion (60 min) and reperfusion (24h), an experimental model in which excessive production of reactive oxygen and nitrogen species has been found. In our data, biochemical analysis of tissue proteins showed that cerebral ischemia/reperfusion (I/R) injury resulted in significant elevation of carbonyl groups and nitrotyrosine in the brain of I/R in comparison to sham controls, indicating the occurrence of oxidative/nitrative modification to brain proteins. HSYA-treatment (1, 5 and 10mg/kg) inhibited I/R-induced protein oxidation and nitration. 12/15-Lipoxygenase (12/15-LOX), the enzyme implicated in oxidative stress of cerebral I/R, displayed overexpression in I/R rats. Elevated 12/15-LOX activity, estimated by the level of its metabolite 15-hydroxyeicosatetraenoic acid (15-HETE), was also induced by the challenge of cerebral I/R. Administration of HSYA 1, 5 and 10mg/kg reduced the upregulation of 12/15-LOX expression and activity in a dose-dependent manner. Moreover, the increase in blood-brain barrier (BBB) permeability evaluated by IgG leakage, Evans blue extravasation, and brain water content, respectively, was markedly alleviated by HSYA, indicating its protection against BBB disruption and brain edema following I/R insult. Taken together, these results demonstrate the neuroprotective properties of HSYA, which may be at least in part due to its potential to reduce protein oxidation and nitration, inhibit the upregulation of 12/15-LOX, and attenuate BBB breakdown.


Subject(s)
Blood-Brain Barrier/drug effects , Chalcone/analogs & derivatives , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Quinones/pharmacology , Reperfusion Injury/drug therapy , Animals , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/metabolism , Blotting, Western , Chalcone/pharmacology , Disease Models, Animal , Fluorescent Antibody Technique , Ischemic Attack, Transient/metabolism , Male , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism
9.
Saudi Med J ; 33(6): 608-16, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22729114

ABSTRACT

OBJECTIVE: To attenuate the effects of early streptozotocin-induced diabetes on renal functions through supplementation with either pravastatin or 12/15-lipoxygenase pathway inhibitors. METHODS: The study was carried out at King Khalid University Hospital, Riyadh, Saudi Arabia from November 2010 to November 2011. Rats were assigned to control rats (group I) receiving vehicle; normoalbuminuric diabetic rats receiving vehicle (group IIa), nordihydroguaiaretic acid (NDGA) (group IIb), NDGA + insulin (group IIc), pravastatin (group IId) or pravastatin + insulin (group IIe); and microalbuminuric diabetic rats receiving vehicle (group IIIa), NDGA (group IIIb), NDGA + insulin (group IIIc), pravastatin (group IIId) or pravastatin + insulin (group IIIe). The NDGA and pravastatin were administered for 4 months. At the end of the experiment, renal function tests were measured and blood samples were analyzed. RESULTS: Both NDGA and pravastatin had favorable effects on renal function to the same extent, and more favorable effects when diabetes was controlled. Indices of diabetic nephropathy (DN) and oxidative stress were reduced by NDGA or pravastatin therapy with no statistical difference between the 2 lines of therapy. CONCLUSION: Pravastatin and 12/15-lipoxygenase pathway inhibitor (NDGA) have beneficial effects on streptozotocin-induced DN. The findings may provide insight into the feasibility of their clinical use as a complementary therapy for the prevention/treatment of DN.


Subject(s)
Anticholesteremic Agents/pharmacology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Pravastatin/pharmacology , Signal Transduction/drug effects , Albuminuria/etiology , Animals , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/drug effects , Blood Glucose/drug effects , Diabetes Mellitus, Experimental , Diabetic Nephropathies/complications , Diabetic Nephropathies/enzymology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Kidney/drug effects , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar , Treatment Outcome
10.
Hepatobiliary Pancreat Dis Int ; 11(2): 193-202, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22484589

ABSTRACT

BACKGROUND: 12-lipoxygenase (12-LOX) has been reported to be an important gene in cancer cell proliferation and survival, and tumor metastasis. However, its role in hepatocellular carcinoma (HCC) cells remains unknown. METHODS: Expression of 12-LOX was assessed in a diethyl-nitrosamine-induced rat HCC model, and in SMMC-7721, HepG2 and L-02 cells using immunohistochemical staining and reverse transcriptase-polymerase chain reaction (RT-PCR). GST-π and Ki-67 were determined in vivo by immunohistochemical staining. Apoptosis was evaluated by TUNEL assay. Cell viability and apoptosis were determined by MTT assay and flow cytometry, respectively. Apoptosis-related proteins in SMMC-7721 and HepG2 cells were detected by Western blotting. RESULTS: Immunohistochemical staining and RT-PCR showed that 12-LOX was over-expressed in rat HCC and two HCC cell lines, while the expression was inhibited by baicalein, a specific inhibitor of 12-LOX. Baicalein inhibited cell proliferation and induced apoptosis in rat HCC and both cell lines in a dose- and time-dependent manner. Our in vivo study demonstrated that baicalein also reduced neoplastic nodules. Mechanistically, baicalein reduced Bcl-2 protein expression coupled with a slight increase of the expression of Bax and activation of caspase-3. Furthermore, baicalein inhibited the activation of ERK-1/2 (phosphorylated). Interestingly, the effects of baicalein were reversed by 12(S)-HETE, a metabolite of 12-LOX. CONCLUSIONS: Inhibition of 12-LOX leads to reduced numbers of HCC cells, partially caused by increased apoptosis. 12-LOX may be a potential molecular target for HCC prevention and treatment.


Subject(s)
Apoptosis/drug effects , Arachidonate 12-Lipoxygenase/drug effects , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Flavanones/pharmacology , Lipoxygenase Inhibitors/pharmacology , Liver Neoplasms/pathology , Animals , Arachidonate 12-Lipoxygenase/metabolism , Carcinoma, Hepatocellular/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Liver Neoplasms/metabolism , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Time Factors , bcl-2-Associated X Protein/metabolism
11.
Wound Repair Regen ; 20(1): 114-22, 2012.
Article in English | MEDLINE | ID: mdl-22211391

ABSTRACT

Cutaneous burn wounds represent a significant public health problem with 500,000 patients per year in the USA seeking medical attention. Immediately after skin burn injury, the volume of the wound burn expands due to a cascade of chemical reactions, including lipid peroxidation chain reactions. Such expansion threatens life and is therefore highly clinically significant. Based on these chemical reactions, the present paper develops for the first time a three-dimensional mathematical model to quantify the propagation of tissue damage within 12 hours post initial burn. We use the model to investigate the effect of supplemental antioxidant vitamin E for intercepting propagation. We show, for example, that if tissue levels of vitamin E tocotrienol are increased, postburn, by five times then this would slow down the lipid peroxide propagation by at least 50%. We chose the alpha-tocotrienol form of vitamin E as it is a potent inhibitor of 12-lipoxygenase, which is known to propagate oxidative lipid damage. Our model is formulated in terms of differential equations, and sensitivity analysis is performed on the parameters to ensure the robustness of the results.


Subject(s)
Antioxidants/pharmacology , Arachidonate 12-Lipoxygenase/drug effects , Burns/metabolism , Lipid Peroxidation , Models, Theoretical , Protective Agents/pharmacology , Tocotrienols/pharmacology , Burns/enzymology , Burns/physiopathology , Enzyme Inhibitors/pharmacology , Humans , Lipid Peroxidation/drug effects , Time Factors , United States
12.
Mol Pharmacol ; 81(3): 420-30, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22155783

ABSTRACT

Platelet activation is important in the regulation of hemostasis and thrombosis. Uncontrolled activation of platelets may lead to arterial thrombosis, which is a major cause of myocardial infarction and stroke. After activation, metabolism of arachidonic acid (AA) by 12-lipoxygenase (12-LOX) may play a significant role in regulating the degree and stability of platelet activation because inhibition of 12-LOX significantly attenuates platelet aggregation in response to various agonists. Protein kinase C (PKC) activation is also known to be an important regulator of platelet activity. Using a newly developed selective inhibitor for 12-LOX and a pan-PKC inhibitor, we investigated the role of PKC in 12-LOX-mediated regulation of agonist signaling in the platelet. To determine the role of PKC within the 12-LOX pathway, a number of biochemical endpoints were measured, including platelet aggregation, calcium mobilization, and integrin activation. Inhibition of 12-LOX or PKC resulted in inhibition of dense granule secretion and attenuation of both aggregation and αIIbß(3) activation. However, activation of PKC downstream of 12-LOX inhibition rescued agonist-induced aggregation and integrin activation. Furthermore, inhibition of 12-LOX had no effect on PKC-mediated aggregation, indicating that 12-LOX is upstream of PKC. These studies support an essential role for PKC downstream of 12-LOX activation in human platelets and suggest 12-LOX as a possible target for antiplatelet therapy.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Platelet Activation , Protein Kinase C/metabolism , Arachidonate 12-Lipoxygenase/drug effects , Blood Platelets/drug effects , Blood Platelets/enzymology , Blood Platelets/metabolism , Blotting, Western , Calcium/metabolism , Enzyme Activation , Flow Cytometry , Gas Chromatography-Mass Spectrometry , Humans , Lipoxygenase Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology
13.
J Med Chem ; 54(15): 5485-97, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21739938

ABSTRACT

We report the discovery of novel small molecule inhibitors of platelet-type 12-human lipoxygenase, which display nanomolar activity against the purified enzyme, using a quantitative high-throughput screen (qHTS) on a library of 153607 compounds. These compounds also exhibit excellent specificity, >50-fold selectivity vs the paralogues, 5-human lipoxygenase, reticulocyte 15-human lipoxygenase type-1, and epithelial 15-human lipoxygenase type-2, and >100-fold selectivity vs ovine cyclooxygenase-1 and human cyclooxygenase-2. Kinetic experiments indicate this chemotype is a noncompetitive inhibitor that does not reduce the active site iron. Moreover, chiral HPLC separation of two of the racemic lead molecules revealed a strong preference for the (-)-enantiomers (IC(50) of 0.43 ± 0.04 and 0.38 ± 0.05 µM) compared to the (+)-enantiomers (IC(50) of >25 µM for both), indicating a fine degree of selectivity in the active site due to chiral geometry. In addition, these compounds demonstrate efficacy in cellular models, which underscores their relevance to disease modification.


Subject(s)
Arachidonate 12-Lipoxygenase/drug effects , Lipoxygenase Inhibitors/pharmacology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/antagonists & inhibitors , Animals , Blood Platelets/enzymology , Humans , Islets of Langerhans/drug effects , Kinetics , Lipoxygenase Inhibitors/chemical synthesis , Lipoxygenase Inhibitors/pharmacokinetics , Mice , Sheep , Stereoisomerism , Structure-Activity Relationship
14.
J Cell Biochem ; 111(2): 266-73, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20506249

ABSTRACT

Acute ethanol intoxication and exposure (AE) has been known to impair wound healing and associated angiogenesis. Here, we found that AE diminished the formation of novel reparative lipid mediator 14S,21-dihydroxy-docosa-4Z,7Z,10Z,12E,16Z,19Z-hexaenoic acid (14S,21-diHDHA) and its biosynthetic intermediate 14S-hydroxy-DHA (14S-HDHA) from docosahexaenoic acid (DHA) in murine wounds. However, AE did not reduce the formation of DHA and the intermediate 21-HDHA. These results indicate that in the biosynthetic pathways of 14S,21-diHDHA in wounds, AE suppresses the 14S-hydroxy-generating activity of 12-lipoxygenase-like (LOX-like), but does not suppress the 21-hydroxy-generating activity of cytochrome P450 and DHA-generating activities. The AE-suppression of 12-LOX-like activity was further confirmed by the diminished formation of 12-hydroxy-eicosatetraenoic acid in wounds under AE. Supplementing 14S,21-diHDHA to wounds rescued the AE-impaired healing and vascularization. 14S,21-diHDHA restored AE-impaired processes of angiogenesis in vitro: endothelial cell migration, tubulogenesis, and phosphorylation of p38 mitogen-activated protein kinase (MAPK). Taken together, the suppression of 14S,21-diHDHA formation is responsible, at least partially, for the AE-impairment of cutaneous wound healing and angiogenesis. Supplementing 14S,21-diHDHA to compensate its deficit in AE-impaired wounds rescues the healing and angiogenesis. These results provide a novel mechanistic insight for AE-impaired wound healing that involves the necessary roles of 14S,21-diHDHA. They also offer leads for developing 14S,21-diHDHA-related therapeutics to ameliorate AE-impairment of wound healing.


Subject(s)
Alcoholic Intoxication/drug therapy , Docosahexaenoic Acids/therapeutic use , Ethanol/adverse effects , Neovascularization, Physiologic/drug effects , Wound Healing/drug effects , Alcoholic Intoxication/complications , Animals , Arachidonate 12-Lipoxygenase/drug effects , Cell Movement , Docosahexaenoic Acids/analogs & derivatives , Docosahexaenoic Acids/pharmacology , Endothelial Cells , Mice , Phosphorylation , p38 Mitogen-Activated Protein Kinases
15.
J Neurosci Res ; 87(9): 1997-2005, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19235890

ABSTRACT

Oxidative mechanisms of injury are important in many neurological disorders. Developing oligodendrocytes (pre-OLs) are particularly sensitive to oxidative stress-mediated injury. We previously demonstrated a novel function of phylloquinone (vitamin K(1)) and menaquinone 4 (MK-4; a major form of vitamin K2) in protecting pre-OLs and immature neurons against glutathione depletion-induced oxidative damage (Li et al. [ 2003] J. Neurosci. 23:5816-5826). Here we report that vitamin K at nanomolar concentrations prevents arachidonic acid-induced oxidative injury to pre-OLs through blocking the activation of 12-lipoxygenase (12-LOX). Arachidonic acid metabolism is a potential source for reactive oxygen species (ROS) generation during ischemia and reperfusion. Exposure of pre-OLs to arachidonic acid resulted in oxidative cell death in a concentration-dependent manner. Administration of vitamin K (K(1) and MK-4) completely prevented the toxicity. Consistent with our previous findings, inhibitors of 12-LOX abolished ROS production and cell death, indicating that activation of 12-LOX is a key event in arachidonic acid-induced pre-OL death. Vitamin K(1) and MK-4 significantly blocked 12-LOX activation and prevented ROS accumulation in pre-OLs challenged with arachidonic acid. However, vitamin K itself did not directly inhibit 12-LOX enzymatic activity when assayed with purified 12-LOX in vitro. These results suggest that vitamin K, or likely its metabolites, acts upstream of activation of 12-LOX in pre-OLs. In summary, our data indicate that vitamin K prevents oxidative cell death by blocking activation of 12-LOX and ROS generation.


Subject(s)
Arachidonate 12-Lipoxygenase/drug effects , Oligodendroglia/drug effects , Oxidative Stress/drug effects , Stem Cells/drug effects , Vitamin K/pharmacology , Animals , Arachidonate 12-Lipoxygenase/metabolism , Arachidonic Acid/antagonists & inhibitors , Arachidonic Acid/biosynthesis , Arachidonic Acid/toxicity , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cytoprotection/drug effects , Cytoprotection/physiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Oligodendroglia/enzymology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Stem Cells/enzymology , Vitamin K/metabolism , Vitamin K 2/analogs & derivatives , Vitamin K 2/metabolism , Vitamin K 2/pharmacology
16.
Neurochem Int ; 53(6-8): 220-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18680775

ABSTRACT

We report, exogenous addition of ceramide significantly increases 12-hydroxyeicosatetraenoic acid [12-(S)-HETE] levels, in a dose-dependent manner. 12-(S)-HETE levels, in 20, 30 and 40microM ceramide exposed rat primary hippocampal cell cultures containing predominantly astrocytes and few neurons and other glial cells (the cultured hippocampal cells were predominantly astrocytes amounting to over 99% of total cells with few neurons and other glial cells) amounted to 207, 260 and 408% of the controls, respectively. However, dihydroceramide, an inactive analog of ceramide did not alter the levels of 12-(S)-HETE. Ceramide also increased the mRNA and protein expression, and activity of 12-lipoxygease (12-LOX) needed for the synthesis of 12(S)-HETE. These results indicate a possible link between ceramide and 12-LOX pathway. However, ceramide did not alter expression of 5-lipoxygenase (5-LOX), another member of the lipoxygenase family. However, ceramide upregulated expression of cytosolic phospholipase-A(2) (cPLA(2)) and cyclooxygenase-2 (COX-2). Further, ceramide caused a significant increase in the levels of reactive oxygen species (ROS). Ceramide-mediated generation of ROS was inhibited by baicalien but not by indomethacin. In addition, ceramide treated cells exhibited increased mRNA expression of DNA damage induced transcript3 (Ddit3). This report which demonstrate induction of pro-carcinogenic 12-LOX pathway by an anticancer ceramide, may be relevant to cancer biologists studying drug resistant tumors and devising potent anticancer therapeutic strategies to treat drug resistant tumors. These results indicate possibility of 12-LOX involvement in ceramide-mediated generation of ROS and cellular oxidative stress. Induction of 12-LOX pathway by ceramide may have implications in understanding pathophysiology of neurodegenerative diseases involving ROS generation and inflammation.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Arachidonate 12-Lipoxygenase/metabolism , Astrocytes/metabolism , Ceramides/pharmacology , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 12-Lipoxygenase/genetics , Astrocytes/drug effects , Cells, Cultured , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Encephalitis/chemically induced , Encephalitis/metabolism , Encephalitis/physiopathology , Hippocampus/drug effects , Hippocampus/metabolism , Oxidative Stress/drug effects , Phospholipases A2/drug effects , Phospholipases A2/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factor CHOP/drug effects , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
17.
Eur J Pharmacol ; 591(1-3): 36-42, 2008 Sep 04.
Article in English | MEDLINE | ID: mdl-18590721

ABSTRACT

The mechanism by which the histone deacetylase (HDAC) inhibitor trichostatin A inhibits epidermal growth factor (EGF)-induced human 12(S)-lipoxygenase expression was studied. Trichostatin A treatment of human epidermoid carcinoma A431 cells inhibited the EGF-induced 12(S)-lipoxygenase enzymatic activity in a dose-dependent manner that was consistent with the expression of 12(S)-lipoxygenase mRNA and protein. Confocal microscopy indicated that trichostatin A treatment of cells resulted in downregulation of EGF-induced c-Jun expression. Western blotting revealed that trichostatin A treatment of cells resulted in downregulation of EGF-induced c-Jun and constitutively Sp1 expression. Results of a chromatin immunoprecipitation assay revealed that trichostatin A treatment of cells also upregulated Sp1 acetylation and attenuated the recruitment of Sp1, c-Jun, and p300 to the 12(S)-lipoxygenase gene promoter. These results suggested that trichostatin A inhibited EGF-induced 12(S)-lipoxygenase expression by multiple mechanisms, including the attenuation of c-Jun and Sp1 expression and p300 recruitment to the 12(S)-lipoxygenase gene promoter.


Subject(s)
Arachidonate 12-Lipoxygenase/drug effects , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Arachidonate 12-Lipoxygenase/metabolism , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Dose-Response Relationship, Drug , E1A-Associated p300 Protein/drug effects , E1A-Associated p300 Protein/metabolism , Enzyme Inhibitors/administration & dosage , Epidermal Growth Factor/pharmacology , Gene Expression Regulation/drug effects , Humans , Hydroxamic Acids/administration & dosage , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-jun/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Sp1 Transcription Factor/drug effects , Sp1 Transcription Factor/metabolism
18.
Stroke ; 39(9): 2538-43, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18635843

ABSTRACT

BACKGROUND AND PURPOSE: The concept of the neurovascular unit suggests that effects on brain vasculature must be considered if neuroprotection is to be achieved in stroke. We previously reported that 12/15-lipoxygenase (12/15-LOX) is upregulated in the peri-infarct area after middle cerebral artery occlusion in mice, and 12/15-LOX contributes to brain damage after ischemia-reperfusion. The current study was designed to investigate 12/15-LOX involvement in vascular injury in the ischemic brain. METHODS: In cell culture, a human brain microvascular endothelial cell line was subjected to either hypoxia or H(2)O(2)-induced oxidative stress with or without lipoxygenase inhibitors. For in vivo studies, mice were subjected to 90 minutes middle cerebral artery occlusion, and the effects of either 12/15-LOX gene knockout or treatment with lipoxygenase inhibitors were compared. Expression of 12/15-LOX and claudin-5 as well as extravasation of immunoglobulin G were detected by immunohistochemistry. Edema was measured as water content of brain hemispheres according to the wet-dry weight method. RESULTS: Brain endothelial cells were protected against hypoxia and H(2)O(2) by the lipoxygenase inhibitor baicalein. After focal ischemia, 12/15-LOX was increased in neurons and endothelial cells. The vascular tight junction protein claudin-5 underwent extensive degradation in the peri-infarct area, which was partially prevented by the lipoxygenase inhibitor baicalein. Leakage of immunoglobulin G into the brain parenchyma was significantly reduced in 12/15-LOX knockout mice as well as wild-type mice treated with baicalein. Likewise, brain edema was significantly ameliorated. CONCLUSIONS: 12/15-LOX may contribute to ischemic brain damage not just by causing neuronal cell death, but also by detrimental effects on the brain microvasculature. 12/15-LOX inhibitors may thus be effective as both neuroprotectants and vasculoprotectants.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Brain Edema/enzymology , Brain Infarction/enzymology , Brain Ischemia/enzymology , Flavanones/therapeutic use , Ischemic Attack, Transient/enzymology , Animals , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/genetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Edema/physiopathology , Brain Edema/prevention & control , Brain Infarction/drug therapy , Brain Infarction/physiopathology , Brain Ischemia/drug therapy , Brain Ischemia/physiopathology , Cells, Cultured , Claudin-5 , Cytoprotection/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Hydrogen Peroxide/pharmacology , Immunoglobulin G/metabolism , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/physiopathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Oxidative Stress/physiology , Tight Junctions/drug effects , Tight Junctions/metabolism
19.
J Neurosci Res ; 86(4): 904-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17960827

ABSTRACT

The lipid-metabolizing enzyme 12/15-lipoxygenase (12/15-LOX) mediates cell death resulting from oxidative stress in both neurons and oligodendrocytes. Specifically, it may contribute to the pathophysiology of stroke and Alzheimer's and Parkinson's diseases. We report here that two of three specific 12/15-LOX inhibitors, derived from a virtual screen by computer modeling and validated by inhibition of recombinant human 15-LOX in vitro, are able to rescue both neuronal as well as oligodendroglial cells from cell death induced by oxidative stress. Thus, in a fairly streamlined process, an initial virtual screen of 50,000 compounds in a library of drug-like molecules has led to the identification of two novel drug candidates for targeting LOX. Future studies of these novel neuroprotective inhibitors of 12/15-LOX may provide new therapeutic opportunities to combat stroke and other neurodegenerative diseases.


Subject(s)
Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/drug effects , Drug Evaluation, Preclinical/methods , Lipoxygenase Inhibitors/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oligodendroglia/drug effects , Animals , Antioxidants/pharmacology , Cells, Cultured , Computer Simulation , Humans , Oxidative Stress/physiology , Rats
20.
J Am Soc Nephrol ; 16(2): 352-62, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15615821

ABSTRACT

Diabetic nephropathy (DN) is characterized by mesangial cell (MC) hypertrophy and progressive accumulation of glomerular extracellular matrix (ECM). It was reported recently that 12/15-lipoxygenase (12/15-LO) expression is increased in high-glucose (HG)-stimulated MC and in experimental DN. 12-LO products could also directly induce MC hypertrophy and ECM expression and mediate growth factor effects, thus implicating the 12/15-LO pathway in DN. Because TGF-beta is a major player in the pathogenesis of DN, whether there is an interplay between the TGF-beta and 12/15-LO pathways in MC was evaluated. Treatment of rat MC (RMC) with TGF-beta significantly increased levels of the 12/15-LO product 12(S)-hydroxyeicosatetraenoic acid [12(S)-HETE] and also 12/15-LO mRNA and protein expression. HG-induced TGF-beta mRNA expression in RMC was inhibited by a specific ribozyme and siRNA targeted to knockdown rat 12/15-LO. It is interesting that direct treatment of RMC with 12(S)-HETE increased TGF-beta mRNA and protein levels, as well as p-Smad2/3, which are TGF-beta-specific target transcription factors. 12(S)-HETE also increased transcription from a minimal TGF-beta promoter. Furthermore, TGF-beta expression and p-Smad2/3 levels were lower in MC from 12/15-LO knockout mice relative to control mice. Reciprocally, mouse MC stably overexpressing 12/15-LO had greater TGF-beta mRNA and also nuclear p-Smad2/3 relative to mock-transfected cells. 12/15-LO and TGF-beta could functionally signal and increase ECM expression via the p38 mitogen-activated protein kinase signaling pathway. These results indicate for the first time that the 12/15-LO and TGF-beta pathways can cross-talk and activate each other. These novel interactions may amplify the signal transduction cascades and molecular events that lead to DN.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Glomerular Mesangium/cytology , Glomerular Mesangium/enzymology , Transforming Growth Factor beta/pharmacology , Animals , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/drug effects , Base Sequence , Blotting, Western , Cell Division/physiology , Cells, Cultured , Mice , Mice, Inbred C57BL , Molecular Sequence Data , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Transfection , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...