Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
JCI Insight ; 7(4)2022 02 22.
Article in English | MEDLINE | ID: mdl-35041620

ABSTRACT

Total body irradiation (TBI) targets sensitive bone marrow hematopoietic cells and gut epithelial cells, causing their death and inducing a state of immunodeficiency combined with intestinal dysbiosis and nonproductive immune responses. We found enhanced Pseudomonas aeruginosa (PAO1) colonization of the gut leading to host cell death and strikingly decreased survival of irradiated mice. The PAO1-driven pathogenic mechanism includes theft-ferroptosis realized via (a) curbing of the host antiferroptotic system, GSH/GPx4, and (b) employing bacterial 15-lipoxygenase to generate proferroptotic signal - 15-hydroperoxy-arachidonoyl-PE (15-HpETE-PE) - in the intestines of irradiated and PAO1-infected mice. Global redox phospholipidomics of the ileum revealed that lysophospholipids and oxidized phospholipids, particularly oxidized phosphatidylethanolamine (PEox), represented the major factors that contributed to the pathogenic changes induced by total body irradiation and infection by PAO1. A lipoxygenase inhibitor, baicalein, significantly attenuated animal lethality, PAO1 colonization, intestinal epithelial cell death, and generation of ferroptotic PEox signals. Opportunistic PAO1 mechanisms included stimulation of the antiinflammatory lipoxin A4, production and suppression of the proinflammatory hepoxilin A3, and leukotriene B4. Unearthing complex PAO1 pathogenic/virulence mechanisms, including effects on the host anti/proinflammatory responses, lipid metabolism, and ferroptotic cell death, points toward potentially new therapeutic and radiomitigative targets.


Subject(s)
Arachidonate 15-Lipoxygenase/genetics , Ferroptosis/genetics , Gene Expression Regulation, Neoplastic , Leukotrienes/genetics , Lipid Peroxides/genetics , Pseudomonas aeruginosa/radiation effects , Radiation Injuries, Experimental/genetics , Animals , Arachidonate 15-Lipoxygenase/biosynthesis , Caco-2 Cells/radiation effects , Female , Humans , Leukotrienes/metabolism , Lipid Peroxides/metabolism , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa/pathogenicity , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology
2.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1866(12): 159042, 2021 12.
Article in English | MEDLINE | ID: mdl-34450267

ABSTRACT

The genome of the cabbage clubroot pathogen Plasmodiophora brassicae Woronin 1877 (Cercozoa, Rhizaria, SAR), possesses two expressed genes encoding the P450s that are phylogenetically related to the enzymes of oxylipin biosynthesis of the CYP74 clan. The cDNA of one of these genes (CYP50918A1) has been expressed in E. coli. The preferred substrate for the recombinant protein, the 13-hydroperoxide of α-linolenic acid (13-HPOT), was converted to the novel heterobicyclic oxylipins, plasmodiophorols A and B (1 and 2) at the ratio ca. 12:1. Compounds 1 and 2 were identified as the substituted 6-oxabicyclo[3.1.0]hexane and 2-oxabicyclo[2.2.1]heptane (respectively) using the MS and NMR spectroscopy, as well as the chemical treatments. The 18O labelling experiments revealed the incorporation of a single 18O atom from [18O2]13-HPOT into the epoxide and ether functions of products 1 and 2 (respectively), but not into their OH groups. In contrast, the 18O from [18O2]water was incorporated only into the hydroxyl functions. One more minor polar product, plasmodiophorol C (3), identified as the cyclopentanediol, was formed through the hydrolysis of compounds 1 and 2. Plasmodiophorols A-C are the congeners of egregiachlorides, hybridalactone, ecklonialactones and related bicyclic oxylipins detected before in some brown and red algae. The mechanism of 13-HPOT conversions to plasmodiophorols A and B involving the epoxyallylic cation intermediate is proposed. The hydroperoxide bicyclase CYP50918A1 is the first enzyme controlling this kind of fatty acid hydroperoxide conversion.


Subject(s)
Lipid Peroxides/genetics , Oxylipins/metabolism , Plasmodiophorida/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Brassica/genetics , Brassica/microbiology , Hydrogen Peroxide/metabolism , Lipid Peroxides/metabolism , Plasmodiophorida/enzymology , Plasmodiophorida/pathogenicity , Prostaglandin-Endoperoxide Synthases/chemistry , Prostaglandin-Endoperoxide Synthases/isolation & purification
3.
J Cell Physiol ; 234(11): 19715-19727, 2019 11.
Article in English | MEDLINE | ID: mdl-30982984

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by pulmonary vascular remodeling of the precapillary pulmonary arteries, with excessive proliferation of vascular cells. This study was performed to examine the effects of long noncoding RNA CPS1 intronic transcript 1 (CPS1-IT) on PAH in rat models of obstructive sleep apnea (OSA) through regulating interleukin (IL)-1ß expression. The OSA models were induced in rats, for determination of the CPS1-IT expression. The binding of CPS1-IT and hypoxia-inducible factor 1 (HIF1) was verified. To analyze the effects of CPS1-IT on PAH, the overexpression vector of CPS1-IT and HIF1, shRNA against IL-1ß and pyrrolidine dithiocarbamate (PDTC, inhibitor of the NF-κB signaling pathway) were injected into rat models, respectively. The blood pressure and activity of biochemical indicators including nitric oxide (NO), nitric oxide synthase (NOS), superoxide dismutase (SOD), and lipid peroxide (LPO) were assessed. The expression of IL-1ß, HIF1, α-smooth muscle actin (α-SMA), proliferating cell nuclear antigen (PCNA), and fibronectin (FN) was determined. The relationship of CPS1-IT to IL-1ß and NF-κB was evaluated. CPS1-IT was downregulated in the OSA rat model. Overexpressed CPS1-IT increased the activity of NO, NOS, and SOD as well as α-SMA expression, whereas decreasing LPO activity and expression of PCNA and FN, whereby PAH was suppressed. Notably, overexpressed CPS1-IT reduced IL-1ß expression through NF-κB signaling pathway via inhibiting the HIF1 transcriptional activity, suggesting a mechanism affecting PAH. To conclude, overexpressed CPS1-IT alleviated PAH in OSA by reducing IL-1ß expression, the mechanism of which was involved with inhibited HIF1 transcriptional activity and the NF-κB signaling pathway.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interleukin-1beta/genetics , Pulmonary Arterial Hypertension/genetics , RNA, Long Noncoding/genetics , Sleep Apnea, Obstructive/genetics , Actins/genetics , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Gene Expression Regulation , Humans , Lipid Peroxides/genetics , NF-kappa B/antagonists & inhibitors , Nitric Oxide/genetics , Nitric Oxide Synthase/genetics , Proliferating Cell Nuclear Antigen/genetics , Proline/analogs & derivatives , Proline/pharmacology , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/pathology , RNA, Small Interfering/genetics , Rats , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/pathology , Superoxide Dismutase/genetics , Thiocarbamates/pharmacology
4.
Proc Natl Acad Sci U S A ; 116(8): 2996-3005, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718432

ABSTRACT

Necroptosis and ferroptosis are two distinct necrotic cell death modalities with no known common molecular mechanisms. Necroptosis is activated by ligands of death receptors such as tumor necrosis factor-α (TNF-α) under caspase-deficient conditions, whereas ferroptosis is mediated by the accumulation of lipid peroxides upon the depletion/or inhibition of glutathione peroxidase 4 (GPX4). The molecular mechanism that mediates the execution of ferroptosis remains unclear. In this study, we identified 2-amino-5-chloro-N,3-dimethylbenzamide (CDDO), a compound known to inhibit heat shock protein 90 (HSP90), as an inhibitor of necroptosis that could also inhibit ferroptosis. We found that HSP90 defined a common regulatory nodal between necroptosis and ferroptosis. We showed that inhibition of HSP90 by CDDO blocked necroptosis by inhibiting the activation of RIPK1 kinase. Furthermore, we showed that the activation of ferroptosis by erastin increased the levels of lysosome-associated membrane protein 2a to promote chaperone-mediated autophagy (CMA), which, in turn, promoted the degradation of GPX4. Importantly, inhibition of CMA stabilized GPX4 and reduced ferroptosis. Our results suggest that activation of CMA is involved in the execution of ferroptosis.


Subject(s)
Autophagy/genetics , Glutathione Peroxidase/genetics , Lysosomal-Associated Membrane Protein 2/genetics , Molecular Chaperones/genetics , Necrosis/genetics , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/drug effects , Caspases/genetics , Cell Death/drug effects , Cell Death/genetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Humans , Iron/metabolism , Ligands , Lipid Peroxides/genetics , Lipid Peroxides/metabolism , Molecular Chaperones/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , Piperazines/metabolism , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/genetics
5.
Acta Ophthalmol ; 91(3): 219-25, 2013 May.
Article in English | MEDLINE | ID: mdl-22429721

ABSTRACT

PURPOSE: Storage time for donor corneas in Optisol GS is limited compared to Eye Bank Organ Culture (EBOC). We here examine the epithelium on donor corneoscleral rims after primary storage in Optisol GS and subsequent incubation in EBOC. METHODS: Morphology was monitored by light and electron microscopy, expression of phenotypic and genotypic markers by immunohistochemistry and RT-PCR and changes in oxidative lipid and DNA damage by ELISA and COMET assay. RESULTS: A prominent loss of cells was observed after storage in Optisol GS. After maintenance in EBOC, spreading apical cells were Occludin(+) , while the staining for E-cadherin and Connexin-43 was less intense. There were an upregulation of Occludin and a downregulation of E-cadherin and Connexin-43. Eye Bank Organ Culture was associated with an ongoing proliferative activity and a downregulation of putative progenitor/stem cell marker ABCG2 and p63. Staining for 8-OHdG and Caspase-3 did not increase, while levels of malondialdehyde and number of DNA strand breaks and oxidized bases increased. CONCLUSIONS: This dual procedure should be pursued as an option to increase the storage time and the pool of available donor corneas. The observed downregulation of markers associated with stemness during EBOC is relevant considering the potential use of donor epithelium in the treatment of ocular surface disorders.


Subject(s)
Chondroitin Sulfates/therapeutic use , Cornea , Cryopreservation/methods , Culture Media, Serum-Free , Dextrans/therapeutic use , Eye Banks/methods , Gentamicins/therapeutic use , Organ Preservation Solutions/therapeutic use , Organ Preservation/methods , Biomarkers/metabolism , Cell Proliferation , Comet Assay , Complex Mixtures/therapeutic use , DNA Damage , Enzyme-Linked Immunosorbent Assay , Epithelium, Corneal/metabolism , Epithelium, Corneal/ultrastructure , Female , Humans , In Situ Nick-End Labeling , Lipid Peroxidation , Lipid Peroxides/genetics , Lipid Peroxides/metabolism , Male , Middle Aged , Organ Culture Techniques , Oxidative Stress , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Donors
6.
J Biol Chem ; 287(21): 17693-17705, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22433871

ABSTRACT

TNFα generates reactive oxygen species (ROS) at the cell surface that induce cell death, but how ROS communicate to mitochondria and their specific apoptotic action(s) are both undefined. ROS oxidize phospholipids to hydroperoxides that are friable and fragment adjacent to the (hydro)peroxide function, forming truncated phospholipids, such as azelaoyl phosphatidylcholine (Az-PC). Az-PC is relatively soluble, and exogenous Az-PC rapidly enters cells to damage mitochondrial integrity and initiate intrinsic apoptosis. We determined whether this toxic phospholipid is formed within cells during TNFα stimulation in sufficient quantities to induce apoptosis and if they are essential in TNFα-induced cytotoxicity. We found that TNFα induced ROS formation and phospholipid peroxidation in Jurkat cells, and either chemical interference with NADPH oxidase activity or siRNA suppression of the NADPH oxidase-4 subunit blocked ROS accumulation and phospholipid peroxidation. Mass spectrometry showed that phospholipid peroxides and then Az-PC increased after TNFα exposure, whereas ROS inhibition abolished Az-PC accumulation and TNFα-induced cell death. Glutathione peroxidase-4 (GPx4), which specifically metabolizes lipid hydroperoxides, fell in TNFα-stimulated cells prior to death. Ectopic GPx4 overcame this, reduced peroxidized phospholipid accumulation, blocked Az-PC accumulation, and prevented death. Conversely, GPx4 siRNA knockdown enhanced phospholipid peroxidation, increasing TNFα-stimulated Az-PC formation and apoptosis. Truncated phospholipids were essential elements of TNFα-induced apoptosis because overexpression of PAFAH2 (a phospholipase A(2) that selectively hydrolyzes truncated phospholipids) blocked TNFα-induced Az-PC accumulation without affecting phospholipid peroxidation. PAFAH2 also abolished apoptosis. Thus, phospholipid oxidation and truncation to apoptotic phospholipids comprise an essential element connecting TNFα receptor signaling to mitochondrial damage and apoptotic death.


Subject(s)
Apoptosis/physiology , Lipid Peroxidation/physiology , Lipid Peroxides/metabolism , Mitochondria/metabolism , Phospholipids/metabolism , Tumor Necrosis Factor-alpha/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Humans , Jurkat Cells , Lipid Peroxides/genetics , Mitochondria/genetics , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , Phospholipids/genetics , Tumor Necrosis Factor-alpha/genetics
7.
J Cancer Res Clin Oncol ; 138(6): 999-1009, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22362301

ABSTRACT

PURPOSE: Breast cancer (BC) is a complex, multi-stage disease involving deregulation of different signaling cascades. The present study was conducted to determine the extent of apoptosis, angiogenesis, inflammation, and oxidative stress in patients with different stages of BC as an approach to disease biological behavior. Therefore, plasma levels of soluble (s) Fas, bcl-2 as antiapoptotic indices; interleukin (IL)-8, tumor necrosis factor (TNF)-α as apoptotic, inflammatory, angiogenic indices; lipid peroxides (LPO), nitric oxide (NO) as oxidative stress and angiogenic indices were measured in patients with BC. METHODS: Thirty-seven newly diagnosed patients with BC, 30 patients with benign breast masses, and 30 healthy controls were recruited. Plasma levels of sFas, bcl-2, IL-8, and TNF-α were measured by immunosorbent assay kits and LPO and NO by chemical methods. RESULTS: Plasma sFas and LPO were significantly higher in BC patients versus benign breast masses and healthy controls (P < 0.0001). Bcl-2, IL-8, TNF-α, and NO were significantly higher in benign breast masses (P < 0.0001, P < 0.037, P < 0.0001, P < 0.001) and BC (P < 0.0001) versus controls and in BC versus benign breast masses (P < 0.0001). sFas, bcl-2, IL-8, TNF-α, LPO, and NO were increased with advanced tumor stages. There were positive correlations between sFas, bcl-2, IL-8 TNF-α, LPO, and NO. CONCLUSIONS: BC tumor cells overexpress bcl-2 and sFas to secure their outgrowth and survival. However, this coincides with activation of physiologic regulatory mechanisms, as increased IL-8, TNF-α, LPO, and NO, which try to stop tumor cells by inducing apoptosis. Outcompeting of these mechanisms result in tumor progression as IL-8, TNF-α, and NO are also angiogenic stimulators.


Subject(s)
Apoptosis/physiology , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Oxidative Stress/physiology , Adult , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cross-Sectional Studies , Female , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-8/blood , Interleukin-8/genetics , Interleukin-8/metabolism , Lipid Peroxides/genetics , Lipid Peroxides/metabolism , Middle Aged , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Nitric Oxide/genetics , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-bcl-2/blood , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/blood , fas Receptor/genetics , fas Receptor/metabolism
8.
J Biol Chem ; 286(30): 26334-43, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21642428

ABSTRACT

Cytochrome c (cyt c) release upon oxidation of cardiolipin (CL) in the mitochondrial inner membrane (IM) under oxidative stress occurs early in the intrinsic apoptotic pathway. We postulated that CL oxidation mobilizes not only cyt c but also CL itself in the form of hydroperoxide (CLOOH) species. Relatively hydrophilic CLOOHs could assist in apoptotic signaling by translocating to the outer membrane (OM), thus promoting recruitment of the pro-apoptotic proteins truncated Bid (tBid) and Bax for generation of cyt c-traversable pores. Initial testing of these possibilities showed that CLOOH-containing liposomes were permeabilized more readily by tBid plus Ca(2+) than CL-containing counterparts. Moreover, CLOOH translocated more rapidly from IM-mimetic to OM-mimetic liposomes than CL and permitted more extensive OM permeabilization. We found that tBid bound more avidly to CLOOH-containing membranes than to CL counterparts, and binding increased with increasing CLOOH content. Permeabilization of CLOOH-containing liposomes in the presence of tBid could be triggered by monomeric Bax, consistent with tBid/Bax cooperation in pore formation. Using CL-null mitochondria from a yeast mutant, we found that tBid binding and cyt c release were dramatically enhanced by transfer acquisition of CLOOH. Additionally, we observed a pre-apoptotic IM-to-OM transfer of oxidized CL in cardiomyocytes treated with the Complex III blocker, antimycin A. These findings provide new mechanistic insights into the role of CL oxidation in the intrinsic pathway of oxidative apoptosis.


Subject(s)
Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Cardiolipins/metabolism , Lipid Peroxides/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , bcl-2-Associated X Protein/metabolism , Animals , BH3 Interacting Domain Death Agonist Protein/genetics , Cardiolipins/genetics , Humans , Lipid Peroxides/genetics , Mice , Mitochondria/genetics , Mutation , Oxidation-Reduction , Permeability , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , bcl-2-Associated X Protein/genetics
9.
J Biol Chem ; 284(1): 46-55, 2009 Jan 02.
Article in English | MEDLINE | ID: mdl-19001413

ABSTRACT

Previously, we demonstrated that mitochondria from denervated muscle exhibited dramatically higher Amplex Red dependent fluorescence (thought to be highly specific for hydrogen peroxide) compared with control muscle mitochondria. We now demonstrate that catalase only partially inhibits the Amplex Red signal in mitochondria from denervated muscle. In contrast, ebselen (a glutathione peroxidase mimetic and inhibitor of fatty acid hydroperoxides) significantly inhibits the Amplex Red signal. This suggests that the majority of the Amplex Red signal in mitochondria from denervated muscle is not derived from hydrogen peroxide. Because Amplex Red cannot react with substrates in the lipid environment, we hypothesize that lipid hydroperoxides formed within the mitochondrial lipid bilayer are released as fatty acid hydroperoxides and react with the Amplex Red probe. We also suggest that the release of fatty acid hydroperoxides from denervated muscle mitochondria may be an important determinant of muscle atrophy. In support of this, muscle atrophy and the Amplex Red signal are inhibited in caloric restricted mice and in transgenic mice that overexpress the lipid hydroperoxide-detoxifying enzyme glutathione peroxidase 4. Finally, we propose that cytosolic phospholipase A2 may be a potential source of these hydroperoxides.


Subject(s)
Lipid Peroxides/biosynthesis , Mitochondria, Muscle/metabolism , Mitochondrial Membranes/metabolism , Muscle, Skeletal/enzymology , Muscular Atrophy/enzymology , Phospholipases A2, Cytosolic/metabolism , Animals , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Lipid Peroxides/genetics , Mice , Mice, Knockout , Mitochondria, Muscle/genetics , Mitochondria, Muscle/pathology , Muscle Denervation , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Phospholipases A2, Cytosolic/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase
10.
Genes Cells ; 12(11): 1235-44, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17986007

ABSTRACT

Idiopathic pulmonary fibrosis (IPF), a chronic progressive lung disease of unknown etiology, is characterized by the expansion of myofibroblasts and abnormal deposition of extracellular matrix in the lung parenchyma. To elucidate the molecular mechanisms that lead to IPF, we analyzed myofibroblasts established from patients with IPF by oligonucleotide microarrays. Gene expression profiles clearly suggested that lipid peroxidation is enhanced in myofibroblasts, which was confirmed by measuring cellular lipid hydroperoxides. One of the most highly up-regulated proteins in myofibroblasts was selenoprotein P, an antioxidant protein not previously associated with IPF. Subsequent analysis demonstrated that selenoprotein P reduces lipid hydroperoxides and maintains the viability of myofibroblasts. Thus, our results reveal a novel pathophysiologic function of myofibroblasts as a generator of lipid hydroperoxides, and a self-defense mechanism against self-generated oxidative stress.


Subject(s)
Fibroblasts/metabolism , Pulmonary Fibrosis/metabolism , Selenoprotein P/metabolism , Cell Survival/physiology , Gene Expression Profiling , Humans , Immunohistochemistry , Lipid Peroxides/genetics , Lipid Peroxides/metabolism , MAP Kinase Kinase 4/metabolism , Oxidation-Reduction , Oxidative Stress , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Selenoprotein P/genetics , Up-Regulation
11.
Brain Res Dev Brain Res ; 133(2): 127-39, 2002 Feb 28.
Article in English | MEDLINE | ID: mdl-11882343

ABSTRACT

Lesch-Nyhan disease, a neurogenetic disorder caused by congenital deficiency of the purine salvage enzyme hypoxanthine guanine phosphoribosyl transferase, is associated with a prominent loss of striatal dopamine. The current studies address the hypothesis that oxidant stress causes damage or dysfunction of nigrostriatal dopamine neurons in a knockout mouse model of the disease, by assessing several markers of oxidative damage and free radical scavenging systems. Some of these measures provided evidence for an increase in oxidative stress in the mutant mice (aconitase activity, oxidized glutathione, and lipid peroxides), but others did not (superoxide dismutase, protein thiol content, carbonyl protein content, total glutathione, glutathione peroxidase, catalase, and thiobarbituric reducing substances). Immunolocalization of heme-oxygenase 1 provided no evidence for oxidative stress restricted to specific elements of the striatum or midbrain in the mutants. Striatal dopamine systems of the mutant mice were more vulnerable to a challenge with the neurotoxin 6-hydroxydopamine, but they were not protected by cross-breeding the mutants with transgenic mice over-expressing superoxide dismutase. Overall, these data provide evidence for increased oxidative stress, but the failure to protect the knockout mice by over-expressing SOD1 argues that oxidative stress is not the sole process responsible for the loss of striatal dopamine.


Subject(s)
Cell Death/genetics , Dopamine/deficiency , Free Radical Scavengers/metabolism , Lesch-Nyhan Syndrome/enzymology , Neostriatum/enzymology , Neurons/enzymology , Oxidative Stress/genetics , 3,4-Dihydroxyphenylacetic Acid/metabolism , Aconitate Hydratase/genetics , Aconitate Hydratase/metabolism , Animals , Disease Models, Animal , Female , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , Lesch-Nyhan Syndrome/genetics , Lesch-Nyhan Syndrome/physiopathology , Lipid Peroxides/genetics , Lipid Peroxides/metabolism , Male , Membrane Proteins , Mice , Mice, Knockout , Mice, Transgenic/physiology , Mutation/physiology , Neostriatum/pathology , Neostriatum/physiopathology , Neurons/pathology , Oxidopamine/pharmacology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
12.
Plant Cell ; 13(3): 613-26, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11251100

ABSTRACT

Plant lipoxygenases (LOXs) are a functionally diverse class of dioxygenases implicated in physiological processes such as growth, senescence, and stress-related responses. LOXs incorporate oxygen into their fatty acid substrates and produce hydroperoxide fatty acids that are precursors of jasmonic acid and related compounds. Here, we report the involvement of the tuber-associated LOXs, designated the Lox1 class, in the control of tuber growth. RNA hybridization analysis showed that the accumulation of Lox1 class transcripts was restricted to developing tubers, stolons, and roots and that mRNA accumulation correlated positively with tuber initiation and growth. In situ hybridization showed that Lox1 class transcripts accumulated in the apical and subapical regions of the newly formed tuber, specifically in the vascular tissue of the perimedullary region, the site of the most active cell growth during tuber enlargement. Suppression mutants produced by expressing antisense coding sequence of a specific tuber LOX, designated POTLX-1, exhibited a significant reduction in LOX activity in stolons and tubers. The suppression of LOX activity correlated with reduced tuber yield, decreased average tuber size, and a disruption of tuber formation. Our results indicate that the pathway initiated by the expression of the Lox1 class genes of potato is involved in the regulation of tuber enlargement.


Subject(s)
Lipoxygenase/biosynthesis , Solanum tuberosum/enzymology , Solanum tuberosum/growth & development , Cell Division , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Plant , Isoenzymes/biosynthesis , Isoenzymes/drug effects , Isoenzymes/genetics , Lipid Peroxides/biosynthesis , Lipid Peroxides/genetics , Lipoxygenase/drug effects , Lipoxygenase/genetics , Phylogeny , Plant Shoots/cytology , Plant Shoots/enzymology , Plant Shoots/genetics , Plant Shoots/growth & development , Plants, Genetically Modified , RNA, Antisense/metabolism , RNA, Plant/genetics , RNA, Plant/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Receptors, Oxidized LDL , Recombinant Proteins , Solanum tuberosum/genetics , Suppression, Genetic , Transcription, Genetic
13.
J Am Soc Nephrol ; 9(11): 1990-7, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9808084

ABSTRACT

Atherogenic lipoproteins such as oxidized LDL are implicated in the pathogenesis of atherosclerosis and renal disease. Fatty acid hydroperoxides and phospholipids such as linoleyl hydroperoxide (LAox or 13-HPODE) and lysophosphatidylcholine (lyso-PC), abundant components of oxidized LDL, mediate the effects of atherogenic lipids. Oxidized LDL has been shown to induce heme oxygenase-1 (HO-1), a microsomal enzyme that is involved in heme detoxification and is a major endogenous source of carbon monoxide. HO-1 is also induced by many other stimuli that shift cellular redox. To identify the constituents and molecular mechanisms of oxidized LDL-mediated HO-1 induction, human renal epithelial cells and aortic endothelial cells were exposed to LAox and lyso-PC. Exposure to LAox (25 microM) showed an approximately 16-fold induction of HO-1 mRNA, whereas exposure to lyso-PC (25 microM) showed only an approximate 2.6-fold increase. Treatment with actinomycin-D (4 microM), a transcriptional inhibitor, as well as nuclear run-on assays, demonstrated that LAox-mediated HO-1 gene induction is dependent on de novo transcription. Cycloheximide did not affect LAox-mediated HO-1 mRNA induction, suggesting that new protein synthesis is not required for transcriptional induction. Transfection of a human HO-1 promoter-reporter gene construct showed that LAox upregulation of HO-1 occurs via mechanisms different from those of known inducers, heme and cadmium. These studies are the first demonstration that LAox induces HO-1 by transcriptional mechanisms and may have implications in the pathogenesis of cell injury in atherosclerosis and progressive renal disease.


Subject(s)
Aorta/physiology , Endothelium, Vascular/physiology , Gene Expression Regulation/physiology , Heme Oxygenase (Decyclizing)/genetics , Kidney/physiology , Linoleic Acids/genetics , Lipid Peroxides/genetics , Transcription, Genetic/physiology , Aorta/cytology , Cells, Cultured , Endothelium, Vascular/cytology , Enzyme Activation/physiology , Epithelial Cells/physiology , Heme Oxygenase-1 , Humans , Kidney/cytology , Lysophosphatidylcholines/pharmacology , Membrane Proteins , RNA, Messenger/biosynthesis , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...