Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Redox Biol ; 58: 102532, 2022 12.
Article En | MEDLINE | ID: mdl-36375379

Currently there are no established therapies to treat high-risk patients with unstable atherosclerotic lesions that are prone to rupture and can result in thrombosis, abrupt arterial occlusion, and a precipitous infarction. Rather than being stenotic, rupture-prone non-occlusive plaques are commonly enriched with inflammatory cells and have a thin fibrous cap. We reported previously that inhibition of the pro-inflammatory enzyme myeloperoxidase (MPO) with the suicide inhibitor AZM198 prevents formation of unstable plaque in the Tandem Stenosis (TS) mouse model of plaque instability. However, in our previous study AZM198 was administered to animals before unstable plaque was present and hence it did not test the significant unmet clinical need present in high-risk patients with vulnerable atherosclerosis. In the present study we therefore asked whether pharmacological inhibition of MPO with AZM198 can stabilize pre-existing unstable lesions in an interventional setting using the mouse model of plaque instability. In vivo molecular magnetic resonance imaging of arterial MPO activity using bis-5-hydroxytryptamide-DTPA-Gd and histological analyses revealed that arterial MPO activity was elevated one week after TS surgery, prior to the presence of unstable lesions observed two weeks after TS surgery. Animals with pre-existing unstable plaque were treated with AZM198 for one or five weeks. Both short- and long-term intervention effectively inhibited arterial MPO activity and increased fibrous cap thickness, indicative of a more stable plaque phenotype. Plaque stabilization was observed without AZM198 affecting the arterial content of Ly6B.2+- and CD68+-cells and MPO protein. These findings demonstrate that inhibition of arterial MPO activity converts unstable into stable atherosclerotic lesions in a preclinical model of plaque instability and highlight the potential therapeutic potency of MPO inhibition for the management of high-risk patients and the development of novel protective strategies against cardiovascular diseases.


Atherosclerosis , Cardiovascular Diseases , Peroxidase , Plaque, Atherosclerotic , Animals , Mice , Atherosclerosis/drug therapy , Cardiovascular Diseases/prevention & control , Disease Models, Animal , Peroxidase/antagonists & inhibitors , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology
2.
Zebrafish ; 19(1): 7-17, 2022 02.
Article En | MEDLINE | ID: mdl-35108124

Heme oxygenase function is highly conserved between vertebrates where it plays important roles in normal embryonic development and controls oxidative stress. Expression of the zebrafish heme oxygenase 1 genes is known to be responsive to oxidative stress suggesting a conserved physiological function. In this study, we generate a knockout allele of zebrafish hmox1a and characterize the effects of hmox1a and hmox1b loss on embryonic development. We find that loss of hmox1a or hmox1b causes developmental defects in only a minority of embryos, in contrast to Hmox1 gene deletions in mice that cause loss of most embryos. Using a tail wound inflammation assay we find a conserved role for hmox1a, but not hmox1b, in normal macrophage migration to the wound site. Together our results indicate that zebrafish hmox1a has clearly a partitioned role from hmox1b that is more consistent with conserved functions of mammalian Heme oxygenase 1.


Heme Oxygenase (Decyclizing) , Zebrafish , Animals , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/pharmacology , Macrophages/metabolism , Mice , Oxidative Stress , Zebrafish/metabolism
3.
Free Radic Biol Med ; 179: 339-362, 2022 02 01.
Article En | MEDLINE | ID: mdl-34775001

Insulin resistance is one of the earliest pathological features of a suite of diseases including type 2 diabetes collectively referred to as metabolic syndrome. There is a growing body of evidence from both pre-clinical studies and human cohorts indicating that reactive oxygen species, such as the superoxide radical anion and hydrogen peroxide are key players in the development of insulin resistance. Here we review the evidence linking mitochondrial reactive oxygen species generated within mitochondria with insulin resistance in adipose tissue and skeletal muscle, two major insulin sensitive tissues. We outline the relevant mitochondria-derived reactive species, how the mitochondrial redox state is regulated, and methodologies available to measure mitochondrial reactive oxygen species. Importantly, we highlight key experimental issues to be considered when studying the role of mitochondrial reactive oxygen species in insulin resistance. Evaluating the available literature on both mitochondrial reactive oxygen species/redox state and insulin resistance in a variety of biological systems, we conclude that the weight of evidence suggests a likely role for mitochondrial reactive oxygen species in the etiology of insulin resistance in adipose tissue and skeletal muscle. However, major limitations in the methods used to study reactive oxygen species in insulin resistance as well as the lack of data linking mitochondrial reactive oxygen species and cytosolic insulin signaling pathways are significant obstacles in proving the mechanistic link between these two processes. We provide a framework to guide future studies to provide stronger mechanistic information on the link between mitochondrial reactive oxygen species and insulin resistance as understanding the source, localization, nature, and quantity of mitochondrial reactive oxygen species, their targets and downstream signaling pathways may pave the way for important new therapeutic strategies.


Diabetes Mellitus, Type 2 , Insulin Resistance , Diabetes Mellitus, Type 2/metabolism , Humans , Mitochondria , Muscle, Skeletal/metabolism , Reactive Oxygen Species/metabolism , Superoxides/metabolism
4.
Nat Commun ; 12(1): 6626, 2021 11 16.
Article En | MEDLINE | ID: mdl-34785665

During systemic inflammation, indoleamine 2,3-dioxygenase 1 (IDO1) becomes expressed in endothelial cells where it uses hydrogen peroxide (H2O2) to oxidize L-tryptophan to the tricyclic hydroperoxide, cis-WOOH, that then relaxes arteries via oxidation of protein kinase G 1α. Here we show that arterial glutathione peroxidases and peroxiredoxins that rapidly eliminate H2O2, have little impact on relaxation of IDO1-expressing arteries, and that purified IDO1 forms cis-WOOH in the presence of peroxiredoxin 2. cis-WOOH oxidizes protein thiols in a selective and stereospecific manner. Compared with its epimer trans-WOOH and H2O2, cis-WOOH reacts slower with the major arterial forms of glutathione peroxidases and peroxiredoxins while it reacts more readily with its target, protein kinase G 1α. Our results indicate a paradigm of redox signaling by H2O2 via its enzymatic conversion to an amino acid-derived hydroperoxide that 'escapes' effective reductive inactivation to engage in selective oxidative activation of key target proteins.


Hydrogen Peroxide/metabolism , Peroxidases/chemistry , Peroxidases/metabolism , Signal Transduction , Animals , Cyclic GMP-Dependent Protein Kinase Type I , Endothelial Cells/metabolism , Homeodomain Proteins/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Peroxidases/genetics , Peroxiredoxins/metabolism , Tryptophan/metabolism
5.
Redox Biol ; 46: 102127, 2021 10.
Article En | MEDLINE | ID: mdl-34521065

Mitochondrial energy production and function rely on optimal concentrations of the essential redox-active lipid, coenzyme Q (CoQ). CoQ deficiency results in mitochondrial dysfunction associated with increased mitochondrial oxidative stress and a range of pathologies. What drives CoQ deficiency in many of these pathologies is unknown, just as there currently is no effective therapeutic strategy to overcome CoQ deficiency in humans. To date, large-scale studies aimed at systematically interrogating endogenous systems that control CoQ biosynthesis and their potential utility to treat disease have not been carried out. Therefore, we developed a quantitative high-throughput method to determine CoQ concentrations in yeast cells. Applying this method to the Yeast Deletion Collection as a genome-wide screen, 30 genes not known previously to regulate cellular concentrations of CoQ were discovered. In combination with untargeted lipidomics and metabolomics, phosphatidylethanolamine N-methyltransferase (PEMT) deficiency was confirmed as a positive regulator of CoQ synthesis, the first identified to date. Mechanistically, PEMT deficiency alters mitochondrial concentrations of one-carbon metabolites, characterized by an increase in the S-adenosylmethionine to S-adenosylhomocysteine (SAM-to-SAH) ratio that reflects mitochondrial methylation capacity, drives CoQ synthesis, and is associated with a decrease in mitochondrial oxidative stress. The newly described regulatory pathway appears evolutionary conserved, as ablation of PEMT using antisense oligonucleotides increases mitochondrial CoQ in mouse-derived adipocytes that translates to improved glucose utilization by these cells, and protection of mice from high-fat diet-induced insulin resistance. Our studies reveal a previously unrecognized relationship between two spatially distinct lipid pathways with potential implications for the treatment of CoQ deficiencies, mitochondrial oxidative stress/dysfunction, and associated diseases.


Mitochondrial Diseases , Ubiquinone , Animals , Genetic Testing , Mice , Mitochondrial Diseases/genetics , Oxidation-Reduction , Phosphatidylethanolamine N-Methyltransferase , Phospholipids , Ubiquinone/metabolism
6.
Nat Protoc ; 16(7): 3382-3418, 2021 07.
Article En | MEDLINE | ID: mdl-34117477

The L-tryptophan-derived tricyclic hydroperoxide cis-WOOH was recently identified as a novel and biologically important factor for regulating vascular tone and blood pressure under inflammatory conditions and potentially other cellular redox signaling events. cis-WOOH is highly labile and currently not available commercially. In this protocol, we provide procedures for the synthesis, purification, quantification and characterization of cis-WOOH, its epimer trans-WOOH and their respective alcohols (cis-WOH and trans-WOH). Photo-oxidation of L-tryptophan (L-Trp) results in a mixture containing cis-WOOH and trans-WOOH, which are separated and purified by semi-preparative HPLC. cis-WOH and trans-WOH are then produced by sodium borohydride reduction and purified by semi-preparative HPLC. Characterization of cis-WOOH and trans-WOOH and the reduced alcohol variants is achieved using HPLC, fluorescence, NMR and liquid chromatography-tandem mass spectrometry. The protocol provides instructions for storage and quantification, as well as ways to test the stability of these hydroperoxides in commonly used buffers and media. Finally, we describe examples of how to monitor the formation of cis-WOOH in biological samples. The protocol ensures reasonable yield (11%) and purity (>99%) of cis-WOOH and control compounds in 5-6 d and outlines conditions under which cis-WOOH is stable for several months.


Hydrogen Peroxide/chemical synthesis , Tryptophan/chemistry , Animals , Chromatography, High Pressure Liquid , Humans , Hydrogen Peroxide/isolation & purification , Mass Spectrometry , Oxidation-Reduction , Reproducibility of Results , Superoxides/chemistry , Temperature
7.
Arterioscler Thromb Vasc Biol ; 41(1): 317-330, 2021 01.
Article En | MEDLINE | ID: mdl-33207934

OBJECTIVE: Hmox1 (heme oxygenase-1) is a stress-induced enzyme that catalyzes the degradation of heme to carbon monoxide, iron, and biliverdin. Induction of Hmox1 and its products protect against cardiovascular disease, including ischemic injury. Hmox1 is also a downstream target of the transcription factor HIF-1α (hypoxia-inducible factor-1α), a key regulator of the body's response to hypoxia. However, the mechanisms by which Hmox1 confers protection against ischemia-mediated injury remain to be fully understood. Approach and Results: Hmox1 deficient (Hmox1-/-) mice had impaired blood flow recovery with severe tissue necrosis and autoamputation following unilateral hindlimb ischemia. Autoamputation preceded the return of blood flow, and bone marrow transfer from littermate wild-type mice failed to prevent tissue injury and autoamputation. In wild-type mice, ischemia-induced expression of Hmox1 in skeletal muscle occurred before stabilization of HIF-1α. Moreover, HIF-1α stabilization and glucose utilization were impaired in Hmox1-/- mice compared with wild-type mice. Experiments exposing dermal fibroblasts to hypoxia (1% O2) recapitulated these key findings. Metabolomics analyses indicated a failure of Hmox1-/- mice to adapt cellular energy reprogramming in response to ischemia. Prolyl-4-hydroxylase inhibition stabilized HIF-1α in Hmox1-/- fibroblasts and ischemic skeletal muscle, decreased tissue necrosis and autoamputation, and restored cellular metabolism to that of wild-type mice. Mechanistic studies showed that carbon monoxide stabilized HIF-1α in Hmox1-/- fibroblasts in response to hypoxia. CONCLUSIONS: Our findings suggest that Hmox1 acts both downstream and upstream of HIF-1α, and that stabilization of HIF-1α contributes to Hmox1's protection against ischemic injury independent of neovascularization.


Heme Oxygenase-1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/enzymology , Membrane Proteins/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/enzymology , Reperfusion Injury/prevention & control , Animals , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Energy Metabolism , Female , Fibroblasts/enzymology , Fibroblasts/pathology , Glucose/metabolism , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Hindlimb , Ischemia/genetics , Ischemia/pathology , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred BALB C , Mice, Knockout , Muscle, Skeletal/pathology , Necrosis , Protein Stability , Regional Blood Flow , Reperfusion Injury/genetics , Reperfusion Injury/pathology
8.
J Biol Chem ; 295(18): 6023-6042, 2020 05 01.
Article En | MEDLINE | ID: mdl-32205446

Coenzyme Q (Q n ) is a vital lipid component of the electron transport chain that functions in cellular energy metabolism and as a membrane antioxidant. In the yeast Saccharomyces cerevisiae, coq1-coq9 deletion mutants are respiratory-incompetent, sensitive to lipid peroxidation stress, and unable to synthesize Q6 The yeast coq10 deletion mutant is also respiratory-deficient and sensitive to lipid peroxidation, yet it continues to produce Q6 at an impaired rate. Thus, Coq10 is required for the function of Q6 in respiration and as an antioxidant and is believed to chaperone Q6 from its site of synthesis to the respiratory complexes. In several fungi, Coq10 is encoded as a fusion polypeptide with Coq11, a recently identified protein of unknown function required for efficient Q6 biosynthesis. Because "fused" proteins are often involved in similar biochemical pathways, here we examined the putative functional relationship between Coq10 and Coq11 in yeast. We used plate growth and Seahorse assays and LC-MS/MS analysis to show that COQ11 deletion rescues respiratory deficiency, sensitivity to lipid peroxidation, and decreased Q6 biosynthesis of the coq10Δ mutant. Additionally, immunoblotting indicated that yeast coq11Δ mutants accumulate increased amounts of certain Coq polypeptides and display a stabilized CoQ synthome. These effects suggest that Coq11 modulates Q6 biosynthesis and that its absence increases mitochondrial Q6 content in the coq10Δcoq11Δ double mutant. This augmented mitochondrial Q6 content counteracts the respiratory deficiency and lipid peroxidation sensitivity phenotypes of the coq10Δ mutant. This study further clarifies the intricate connection between Q6 biosynthesis, trafficking, and function in mitochondrial metabolism.


Gene Deletion , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Ubiquinone/analogs & derivatives , Gene Expression Regulation, Fungal , Gene Knockout Techniques , Humans , Mitochondria/metabolism , Protein Transport , Saccharomyces cerevisiae/metabolism , Ubiquinone/biosynthesis , Ubiquinone/deficiency , Ubiquinone/genetics , Ubiquinone/metabolism
10.
J Biol Chem ; 295(4): 981-993, 2020 01 24.
Article En | MEDLINE | ID: mdl-31826918

Ubiquinone 8 (coenzyme Q8 or Q8) mediates electron transfer within the aerobic respiratory chain, mitigates oxidative stress, and contributes to gene expression in Escherichia coli In addition, Q8 was proposed to confer bacterial osmotolerance by accumulating during growth at high osmotic pressure and altering membrane stability. The osmolyte trehalose and membrane lipid cardiolipin accumulate in E. coli cells cultivated at high osmotic pressure. Here, Q8 deficiency impaired E. coli growth at low osmotic pressure and rendered growth osmotically sensitive. The Q8 deficiency impeded cellular O2 uptake and also inhibited the activities of two proton symporters, the osmosensing transporter ProP and the lactose transporter LacY. Q8 supplementation decreased membrane fluidity in liposomes, but did not affect ProP activity in proteoliposomes, which is respiration-independent. Liposomes and proteoliposomes prepared with E. coli lipids were used for these experiments. Similar oxygen uptake rates were observed for bacteria cultivated at low and high osmotic pressures. In contrast, respiration was dramatically inhibited when bacteria grown at the same low osmotic pressure were shifted to high osmotic pressure. Thus, respiration was restored during prolonged growth of E. coli at high osmotic pressure. Of note, bacteria cultivated at low and high osmotic pressures had similar Q8 concentrations. The protection of respiration was neither diminished by cardiolipin deficiency nor conferred by trehalose overproduction during growth at low osmotic pressure, but rather might be achieved by Q8-independent respiratory chain remodeling. We conclude that osmotolerance is conferred through Q8-independent protection of respiration, not by altering physical properties of the membrane.


Escherichia coli/growth & development , Osmotic Pressure , Ubiquinone/pharmacology , Aerobiosis/drug effects , Anisotropy , Escherichia coli/drug effects , Escherichia coli Proteins/metabolism , Fluorescence , Membrane Fluidity/drug effects , Membrane Transport Proteins/metabolism , Mutation/genetics , Osmolar Concentration , Proteolipids/metabolism , Trehalose/metabolism
11.
JCI Insight ; 4(21)2019 11 01.
Article En | MEDLINE | ID: mdl-31581149

The RBC storage lesion is a multiparametric response that occurs during storage at 4°C, but its impact on transfused patients remains unclear. In studies of the RBC storage lesion, the temperature transition from cold storage to normal body temperature that occurs during transfusion has received limited attention. We hypothesized that multiple deleterious events might occur in this period of increasing temperature. We show dramatic alterations in several properties of therapeutic blood units stored at 4°C after warming them to normal body temperature (37°C), as well as febrile temperature (40°C). In particular, the intracellular content and redox state of NADP(H) were directly affected by post-storage incubation at 37°C, as well as by pro-oxidant storage conditions. Modulation of the NADPH-producing pentose phosphate pathway, but not the prevention of hemoglobin autoxidation by conversion of oxyhemoglobin to carboxyhemoglobin, provided protection against storage-induced alterations in RBCs, demonstrating the central role of NADPH in mitigating increased susceptibility of stored RBCs to oxidative stress. We propose that assessing RBC oxidative status after restoration of body temperature constitutes a sensitive method for detecting storage-related alterations that has the potential to improve the quality of stored RBCs for transfusion.


Erythrocytes/metabolism , Hot Temperature , NADP/metabolism , Oxidative Stress , Adult , Female , Humans , Inosine/administration & dosage , Male , Middle Aged , Pyruvic Acid/administration & dosage
12.
Elife ; 82019 03 26.
Article En | MEDLINE | ID: mdl-30912741

ATP synthase produces the majority of cellular energy in most cells. We have previously reported cryo-EM maps of autoinhibited E. coli ATP synthase imaged without addition of nucleotide (Sobti et al. 2016), indicating that the subunit ε engages the α, ß and γ subunits to lock the enzyme and prevent functional rotation. Here we present multiple cryo-EM reconstructions of the enzyme frozen after the addition of MgATP to identify the changes that occur when this ε inhibition is removed. The maps generated show that, after exposure to MgATP, E. coli ATP synthase adopts a different conformation with a catalytic subunit changing conformation substantially and the ε C-terminal domain transitioning via an intermediate 'half-up' state to a condensed 'down' state. This work provides direct evidence for unique conformational states that occur in E. coli ATP synthase when ATP binding prevents the ε C-terminal domain from entering the inhibitory 'up' state.


Adenosine Triphosphate/metabolism , Escherichia coli Proteins/ultrastructure , Mitochondrial Proton-Translocating ATPases/ultrastructure , Cryoelectron Microscopy , Protein Conformation , Protein Subunits/chemistry
13.
Nature ; 566(7745): 548-552, 2019 02.
Article En | MEDLINE | ID: mdl-30760924

Singlet molecular oxygen (1O2) has well-established roles in photosynthetic plants, bacteria and fungi1-3, but not in mammals. Chemically generated 1O2 oxidizes the amino acid tryptophan to precursors of a key metabolite called N-formylkynurenine4, whereas enzymatic oxidation of tryptophan to N-formylkynurenine is catalysed by a family of dioxygenases, including indoleamine 2,3-dioxygenase 15. Under inflammatory conditions, this haem-containing enzyme is expressed in arterial endothelial cells, where it contributes to the regulation of blood pressure6. However, whether indoleamine 2,3-dioxygenase 1 forms 1O2 and whether this contributes to blood pressure control have remained unknown. Here we show that arterial indoleamine 2,3-dioxygenase 1 regulates blood pressure via formation of 1O2. We observed that in the presence of hydrogen peroxide, the enzyme generates 1O2 and that this is associated with the stereoselective oxidation of L-tryptophan to a tricyclic hydroperoxide via a previously unrecognized oxidative activation of the dioxygenase activity. The tryptophan-derived hydroperoxide acts in vivo as a signalling molecule, inducing arterial relaxation and decreasing blood pressure; this activity is dependent on Cys42 of protein kinase G1α. Our findings demonstrate a pathophysiological role for 1O2 in mammals through formation of an amino acid-derived hydroperoxide that regulates vascular tone and blood pressure under inflammatory conditions.


Blood Pressure/physiology , Inflammation/blood , Inflammation/physiopathology , Singlet Oxygen/metabolism , Vasodilator Agents/metabolism , Animals , Cell Line , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/chemistry , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cysteine/metabolism , Enzyme Activation/drug effects , Female , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation/enzymology , Male , Oxidation-Reduction/drug effects , Rats , Signal Transduction , Singlet Oxygen/chemistry , Tryptophan/chemistry , Tryptophan/metabolism
14.
Free Radic Biol Med ; 115: 156-165, 2018 02 01.
Article En | MEDLINE | ID: mdl-29195835

Bilirubin, a byproduct of heme catabolism, has been shown to be an effective lipid-soluble antioxidant in vitro. Bilirubin is able to inhibit free radical chain reactions and protects against oxidant-induced damage in vitro and ex vivo. However, direct evidence for bilirubin's antioxidant effects in vivo remains limited. As bilirubin is formed from biliverdin by biliverdin reductase, we generated global biliverdin reductase-a gene knockout (Bvra-/-) mice to assess the contribution of bilirubin as an endogenous antioxidant. Bvra-/- mice appear normal and are born at the expected Mendelian ratio from Bvra+/- x Bvra+/- matings. Compared with corresponding littermate Bvra+/+ and Bvra+/- animals, Bvra-/- mice have green gall bladders and their plasma concentrations of biliverdin and bilirubin are approximately 25-fold higher and 100-fold lower, respectively. Naïve Bvra-/- and Bvra+/+ mice have comparable plasma lipid profiles and low-molecular weight antioxidants, i.e., ascorbic acid, α-tocopherol and ubiquinol-9. Compared with wild-type littermates, however, plasma from Bvra-/- mice contains higher concentrations of cholesteryl ester hydroperoxides (CE-OOH), and their peroxiredoxin 2 (Prx2) in erythrocytes is more oxidized as assessed by the extent of Prx2 dimerization. These data show that Bvra-/- mice experience higher oxidative stress in blood, implying that plasma bilirubin attenuates endogenous oxidative stress.


Cholesterol Esters/metabolism , Erythrocytes/physiology , Heme/metabolism , Oxidative Stress/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Peroxiredoxins/metabolism , Animals , Antioxidants/metabolism , Bilirubin/metabolism , Biliverdine/metabolism , Dimerization , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Sequence Deletion/genetics
15.
Physiol Rev ; 96(4): 1449-508, 2016 10.
Article En | MEDLINE | ID: mdl-27604527

Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. While initially considered to be waste products, CO and biliverdin/bilirubin have been shown over the last 20 years to modulate key cellular processes, such as inflammation, cell proliferation, and apoptosis, as well as antioxidant defense. This shift in paradigm has led to the importance of heme oxygenases and their products in cell physiology now being well accepted. The identification of the two human cases thus far of heme oxygenase deficiency and the generation of mice deficient in Hmox1 or Hmox2 have reiterated a role for these enzymes in both normal cell function and disease pathogenesis, especially in the context of cardiovascular disease. This review covers the current knowledge on the function of both Hmox1 and Hmox2 at both a cellular and tissue level in the cardiovascular system. Initially, the roles of heme oxygenases in vascular health and the regulation of processes central to vascular diseases are outlined, followed by an evaluation of the role(s) of Hmox1 and Hmox2 in various diseases such as atherosclerosis, intimal hyperplasia, myocardial infarction, and angiogenesis. Finally, the therapeutic potential of heme oxygenases and their products are examined in a cardiovascular disease context, with a focus on how the knowledge we have gained on these enzymes may be capitalized in future clinical studies.


Cardiovascular Diseases/enzymology , Cardiovascular System/enzymology , Heme Oxygenase (Decyclizing)/metabolism , Heme/metabolism , Animals , Biliverdine/metabolism , Carbon Monoxide/metabolism , Humans , Iron/metabolism
16.
Free Radic Biol Med ; 87: 137-47, 2015 Oct.
Article En | MEDLINE | ID: mdl-25937176

Diabetes-induced cardiac complications include left ventricular (LV) dysfunction and heart failure. We previously demonstrated that LV phosphoinositide 3-kinase p110α (PI3K) protects the heart against diabetic cardiomyopathy, associated with reduced NADPH oxidase expression and activity. Conversely, in dominant negative PI3K(p110α) transgenic mice (dnPI3K), reduced cardiac PI3K signaling exaggerated diabetes-induced cardiomyopathy, associated with upregulated NADPH oxidase. The goal was to examine whether chronic supplementation with the antioxidant coenzyme Q(10) (CoQ(10)) could attenuate LV superoxide and diabetic cardiomyopathy in a setting of impaired PI3K signaling. Diabetes was induced in 6-week-old nontransgenic and dnPI3K male mice via streptozotocin. After 4 weeks of diabetes, CoQ(10) supplementation commenced (10 mg/kg ip, 3 times/week, 8 weeks). At study end (12 weeks of diabetes), markers of LV function, cardiomyocyte hypertrophy, collagen deposition, NADPH oxidase, oxidative stress (3-nitrotyrosine), and concentrations of CoQ(9) and CoQ(10) were determined. LV NADPH oxidase (Nox2 gene expression and activity, and lucigenin-enhanced chemiluminescence), as well as oxidative stress, were increased by diabetes, exaggerated in diabetic dnPI3K mice, and attenuated by CoQ(10). Diabetes-induced LV diastolic dysfunction (prolonged deceleration time, elevated end-diastolic pressure, impaired E/A ratio), cardiomyocyte hypertrophy and fibrosis, expression of atrial natriuretic peptide, connective tissue growth factor, and ß-myosin heavy chain were all attenuated by CoQ(10). Chronic CoQ(10) supplementation attenuates aspects of diabetic cardiomyopathy, even in a setting of reduced cardiac PI3K protective signaling. Given that CoQ(10) supplementation has been suggested to have positive outcomes in heart failure patients, chronic CoQ(10) supplementation may be an attractive adjunct therapy for diabetic heart failure.


Class I Phosphatidylinositol 3-Kinases/biosynthesis , Diabetic Cardiomyopathies/drug therapy , Oxidative Stress/drug effects , Ubiquinone/analogs & derivatives , Ventricular Dysfunction, Left/drug therapy , Animals , Antioxidants/administration & dosage , Class I Phosphatidylinositol 3-Kinases/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Humans , Male , Mice , Mice, Transgenic , NADPH Oxidases/biosynthesis , NADPH Oxidases/genetics , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/drug effects , Ubiquinone/administration & dosage , Ubiquinone/metabolism , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology
17.
Annu Rev Nutr ; 35: 175-213, 2015.
Article En | MEDLINE | ID: mdl-25974695

Coenzyme Q (CoQ) is an essential lipid of cells present in all cellular compartments. The functions of CoQ in mitochondrial respiration and as an antioxidant are established, although the lipid likely has additional, presently unknown, roles. While the therapeutic utility of CoQ10 supplements is recognized in the rare cases of primary CoQ10 deficiencies, a potential role for CoQ10 supplements in cardiovascular disease, particularly heart failure, has also been studied for over 40 years. This review summarizes our current knowledge in these areas derived from animal studies and human trials. Current evidence for a benefit of CoQ10 supplements in diseases other than primary CoQ10 deficiencies is insufficient.


Heart Failure , Myocardial Ischemia , Ubiquinone/analogs & derivatives , Age Factors , Animals , Antioxidants , Diet , Dietary Supplements , Heart Failure/drug therapy , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle, Skeletal , Muscular Diseases/chemically induced , Muscular Diseases/prevention & control , Myocardial Ischemia/drug therapy , Tissue Distribution , Ubiquinone/administration & dosage , Ubiquinone/deficiency , Ubiquinone/physiology
18.
FEMS Yeast Res ; 14(1): 60-72, 2014 Feb.
Article En | MEDLINE | ID: mdl-24164795

Ageing cells undergo changes in redox homeostasis and acquire high levels of reactive oxygen species (ROS). Because accumulation of ROS involves a change in redox state of cells, functions that are involved in setting redox and maintaining redox homeostasis are very relevant to an understanding of the possible roles of redox homeostasis and ROS in ageing. This review discusses these aspects of ROS in relation to replicative ageing in the model organism Saccharomyces cerevisiae, with reference to ROS generated in cells; cellular responses to oxidative stress; and how cells maintain redox homeostasis in different cellular compartments. It also considers when ROS generation begins as cells age, which ROS species are relevant to ageing and which cellular compartments and processes may contribute ROS to the ageing process. The discussion also covers the heterogeneity of cells with respect to ROS accumulation at particular cell ages, and the possibility of testing the oxidative theory of ageing in yeast cells.


Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/physiology , Homeostasis , Models, Biological , Oxidation-Reduction , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism
19.
G3 (Bethesda) ; 3(12): 2321-33, 2013 Dec 09.
Article En | MEDLINE | ID: mdl-24142923

The heat-shock response in cells, involving increased transcription of a specific set of genes in response to a sudden increase in temperature, is a highly conserved biological response occurring in all organisms. Despite considerable attention to the processes activated during heat shock, less is known about the role of genes in survival of a sudden temperature increase. Saccharomyces cerevisiae genes involved in the maintenance of heat-shock resistance in exponential and stationary phase were identified by screening the homozygous diploid deletants in nonessential genes and the heterozygous diploid mutants in essential genes for survival after a sudden shift in temperature from 30 to 50°. More than a thousand genes were identified that led to altered sensitivity to heat shock, with little overlap between them and those previously identified to affect thermotolerance. There was also little overlap with genes that are activated or repressed during heat-shock, with only 5% of them regulated by the heat-shock transcription factor. The target of rapamycin and protein kinase A pathways, lipid metabolism, vacuolar H(+)-ATPase, vacuolar protein sorting, and mitochondrial genome maintenance/translation were critical to maintenance of resistance. Mutants affected in l-tryptophan metabolism were heat-shock resistant in both growth phases; those affected in cytoplasmic ribosome biogenesis and DNA double-strand break repair were resistant in stationary phase, and in mRNA catabolic processes in exponential phase. Mutations affecting mitochondrial genome maintenance were highly represented in sensitive mutants. The cell division transcription factor Swi6p and Hac1p involved in the unfolded protein response also play roles in maintenance of heat-shock resistance.


Heat-Shock Response/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/physiology , Basic-Leucine Zipper Transcription Factors/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Repair/genetics , Gene Expression Regulation, Fungal , Genome, Fungal , Heterozygote , Mutation , Protein Transport , RNA, Messenger/metabolism , Repressor Proteins/genetics , Reproducibility of Results , Ribosomes/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomere , Transcription Factors/genetics , Tryptophan/genetics , Tryptophan/metabolism
20.
PLoS One ; 8(6): e65240, 2013.
Article En | MEDLINE | ID: mdl-23762325

Responses to many growth and stress conditions are assumed to act via changes to the cellular redox status. However, direct measurement of pH-adjusted redox state during growth and stress has never been carried out. Organellar redox state (E GSH) was measured using the fluorescent probes roGFP2 and pHluorin in Saccharomyces cerevisiae. In particular, we investigated changes in organellar redox state in response to various growth and stress conditions to better understand the relationship between redox-, oxidative- and environmental stress response systems. E GSH values of the cytosol, mitochondrial matrix and peroxisome were determined in exponential and stationary phase in various media. These values (-340 to -350 mV) were more reducing than previously reported. Interestingly, sub-cellular redox state remained unchanged when cells were challenged with stresses previously reported to affect redox homeostasis. Only hydrogen peroxide and heat stress significantly altered organellar redox state. Hydrogen peroxide stress altered the redox state of the glutathione disulfide/glutathione couple (GSSG, 2H(+)/2GSH) and pH. Recovery from moderate hydrogen peroxide stress was most rapid in the cytosol, followed by the mitochondrial matrix, with the peroxisome the least able to recover. Conversely, the bulk of the redox shift observed during heat stress resulted from alterations in pH and not the GSSG, 2H(+)/2GSH couple. This study presents the first direct measurement of pH-adjusted redox state in sub-cellular compartments during growth and stress conditions. Redox state is distinctly regulated in organelles and data presented challenge the notion that perturbation of redox state is central in the response to many stress conditions.


Cell Compartmentation , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Stress, Physiological , Adaptation, Physiological/drug effects , Carbon/pharmacology , Cell Compartmentation/drug effects , Cytosol/drug effects , Cytosol/metabolism , Fermentation/drug effects , Green Fluorescent Proteins/metabolism , Heat-Shock Response/drug effects , Hydrogen Peroxide/toxicity , Hydrogen-Ion Concentration/drug effects , Microscopy, Confocal , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Paraquat/toxicity , Peroxisomes/drug effects , Peroxisomes/metabolism , Potassium/pharmacology , Reproducibility of Results , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development , Sodium/pharmacology , Sorbitol/pharmacology , Stress, Physiological/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
...