Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 11(12): e2306469, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38235614

ABSTRACT

In Alzheimer's disease (AD), dysfunctional mitochondrial metabolism is associated with synaptic loss, the major pathological correlate of cognitive decline. Mechanistic insight for this relationship, however, is still lacking. Here, comparing isogenic wild-type and AD mutant human induced pluripotent stem cell (hiPSC)-derived cerebrocortical neurons (hiN), evidence is found for compromised mitochondrial energy in AD using the Seahorse platform to analyze glycolysis and oxidative phosphorylation (OXPHOS). Isotope-labeled metabolic flux experiments revealed a major block in activity in the tricarboxylic acid (TCA) cycle at the α-ketoglutarate dehydrogenase (αKGDH)/succinyl coenzyme-A synthetase step, metabolizing α-ketoglutarate to succinate. Associated with this block, aberrant protein S-nitrosylation of αKGDH subunits inhibited their enzyme function. This aberrant S-nitrosylation is documented not only in AD-hiN but also in postmortem human AD brains versus controls, as assessed by two separate unbiased mass spectrometry platforms using both SNOTRAP identification of S-nitrosothiols and chemoselective-enrichment of S-nitrosoproteins. Treatment with dimethyl succinate, a cell-permeable derivative of a TCA substrate downstream to the block, resulted in partial rescue of mitochondrial bioenergetic function as well as reversal of synapse loss in AD-hiN. These findings have therapeutic implications that rescue of mitochondrial energy metabolism can ameliorate synaptic loss in hiPSC-based models of AD.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Humans , Alzheimer Disease/metabolism , Induced Pluripotent Stem Cells/metabolism , Energy Metabolism/physiology , Glycolysis , Neurons/metabolism
2.
Cell Chem Biol ; 30(8): 965-975.e6, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37478858

ABSTRACT

A causal relationship between mitochondrial metabolic dysfunction and neurodegeneration has been implicated in synucleinopathies, including Parkinson disease (PD) and Lewy body dementia (LBD), but underlying mechanisms are not fully understood. Here, using human induced pluripotent stem cell (hiPSC)-derived neurons with mutation in the gene encoding α-synuclein (αSyn), we report the presence of aberrantly S-nitrosylated proteins, including tricarboxylic acid (TCA) cycle enzymes, resulting in activity inhibition assessed by carbon-labeled metabolic flux experiments. This inhibition principally affects α-ketoglutarate dehydrogenase/succinyl coenzyme-A synthetase, metabolizing α-ketoglutarate to succinate. Notably, human LBD brain manifests a similar pattern of aberrantly S-nitrosylated TCA enzymes, indicating the pathophysiological relevance of these results. Inhibition of mitochondrial energy metabolism in neurons is known to compromise dendritic length and synaptic integrity, eventually leading to neuronal cell death. Our evidence indicates that aberrant S-nitrosylation of TCA cycle enzymes contributes to this bioenergetic failure.


Subject(s)
Induced Pluripotent Stem Cells , Parkinson Disease , Synucleinopathies , Humans , Synucleinopathies/metabolism , Induced Pluripotent Stem Cells/metabolism , Parkinson Disease/metabolism , Neurons/metabolism , Brain/metabolism
3.
J Biol Chem ; 298(8): 102159, 2022 08.
Article in English | MEDLINE | ID: mdl-35750212

ABSTRACT

Lysosomal storage diseases result in various developmental and physiological complications, including cachexia. To study the causes for the negative energy balance associated with cachexia, we assessed the impact of sulfamidase deficiency and heparan sulfate storage on energy homeostasis and metabolism in a mouse model of type IIIa mucopolysaccharidosis (MPS IIIa, Sanfilippo A syndrome). At 12-weeks of age, MPS IIIa mice exhibited fasting and postprandial hypertriglyceridemia compared with wildtype mice, with a reduction of white and brown adipose tissues. Partitioning of dietary [3H]triolein showed a marked increase in intestinal uptake and secretion, whereas hepatic production and clearance of triglyceride-rich lipoproteins did not differ from wildtype controls. Uptake of dietary triolein was also elevated in brown adipose tissue (BAT), and notable increases in beige adipose tissue occurred, resulting in hyperthermia, hyperphagia, hyperdipsia, and increased energy expenditure. Furthermore, fasted MPS IIIa mice remained hyperthermic when subjected to low temperature but became cachexic and profoundly hypothermic when treated with a lipolytic inhibitor. We demonstrated that the reliance on increased lipid fueling of BAT was driven by a reduced ability to generate energy from stored lipids within the depot. These alterations arose from impaired autophagosome-lysosome fusion, resulting in increased mitochondria content in beige and BAT. Finally, we show that increased mitochondria content in BAT and postprandial dyslipidemia was partially reversed upon 5-week treatment with recombinant sulfamidase. We hypothesize that increased BAT activity and persistent increases in energy demand in MPS IIIa mice contribute to the negative energy balance observed in patients with MPS IIIa.


Subject(s)
Hypertriglyceridemia , Mucopolysaccharidosis III , Adipose Tissue, Brown/metabolism , Animals , Cachexia , Mice , Mitophagy , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/therapy , Triolein
4.
JCI Insight ; 6(20)2021 10 22.
Article in English | MEDLINE | ID: mdl-34676827

ABSTRACT

Macrophage proinflammatory activation is an important etiologic component of the development of insulin resistance and metabolic dysfunction in obesity. However, the underlying mechanisms are not clearly understood. Here, we demonstrate that a mitochondrial inner membrane protein, adenine nucleotide translocase 2 (ANT2), mediates proinflammatory activation of adipose tissue macrophages (ATMs) in obesity. Ant2 expression was increased in ATMs of obese mice compared with lean mice. Myeloid-specific ANT2-knockout (ANT2-MKO) mice showed decreased adipose tissue inflammation and improved insulin sensitivity and glucose tolerance in HFD/obesity. At the molecular level, we found that ANT2 mediates free fatty acid-induced mitochondrial permeability transition, leading to increased mitochondrial reactive oxygen species production and damage. In turn, this increased HIF-1α expression and NF-κB activation, leading to proinflammatory macrophage activation. Our results provide a previously unknown mechanism for how obesity induces proinflammatory activation of macrophages with propagation of low-grade chronic inflammation (metaflammation).


Subject(s)
Adenine Nucleotide Translocator 2/metabolism , Inflammation/genetics , Macrophage Activation/genetics , Obesity/genetics , Animals , Disease Models, Animal , Female , Humans , Male , Mice
5.
EMBO J ; 40(16): e107901, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34169542

ABSTRACT

How natural or innate-like lymphocytes generate the capacity to produce IL-4 and other cytokines characteristic of type 2 immunity remains unknown. Invariant natural killer T (iNKT) cells differentiate in the thymus into NKT1, NKT2, and NKT17 subsets, similar to mature, peripheral CD4+ T helper cells. The mechanism for this differentiation was not fully understood. Here, we show that NKT2 cells required higher and prolonged calcium (Ca2+ ) signals and continuing activity of the calcium release-activated calcium (CRAC) channel, than their NKT1 counterparts. The sustained Ca2+ entry via CRAC pathway in NKT2 cells was apparently mediated by ORAI and controlled in part by the large mitochondrial Ca2+ uptake. Unique properties of mitochondria in NKT2 cells, including high activity of oxidative phosphorylation, may regulate mitochondrial Ca2+ buffering in NKT2 cells. In addition, the low Ca2+ extrusion rate may also contribute to the higher Ca2+ level in NKT2 cells. Altogether, we identified ORAI-dependent Ca2+ signaling connected with mitochondria and cellular metabolism, as a central regulatory pathway for the differentiation of NKT2 cells.


Subject(s)
Calcium/metabolism , Cell Differentiation , Natural Killer T-Cells/metabolism , Thymus Gland/cytology , Animals , Calcium Release Activated Calcium Channels/metabolism , Calcium Signaling , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/metabolism
6.
J Immunol ; 206(6): 1181-1193, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33547171

ABSTRACT

CCR6+CXCR3+CCR4-CD4+ memory T cells, termed Th1*, are important for long-term immunity to Mycobacterium tuberculosis and the pathogenesis of autoimmune diseases. Th1* cells express a unique set of lineage-specific transcription factors characteristic of both Th1 and Th17 cells and display distinct gene expression profiles compared with other CD4+ T cell subsets. To examine molecules and signaling pathways important for the effector function of Th1* cells, we performed loss-of-function screening of genes selectively enriched in the Th1* subset. The genetic screen yielded candidates whose depletion significantly impaired TCR-induced IFN-γ production. These included genes previously linked to IFN-γ or M. tuberculosis susceptibility and novel candidates, such as ISOC1, encoding a metabolic enzyme of unknown function in mammalian cells. ISOC1-depleted T cells, which produced less IFN-γ and IL-17, displayed defects in oxidative phosphorylation and glycolysis and impairment of pyrimidine metabolic pathway. Supplementation with extracellular pyrimidines rescued both bioenergetics and IFN-γ production in ISOC1-deficient T cells, indicating that pyrimidine metabolism is a key driver of effector functions in CD4+ T cells and Th1* cells. Results provide new insights into the immune-stimulatory function of ISOC1 as well as the particular metabolic requirements of human memory T cells, providing a novel resource for understanding long-term T cell-driven responses.


Subject(s)
Hydrolases/metabolism , Interferon-gamma/genetics , Interleukin-17/genetics , Th1 Cells/immunology , Gene Expression Regulation/immunology , Gene Knockdown Techniques , HEK293 Cells , Healthy Volunteers , Humans , Hydrolases/genetics , Immunologic Memory/genetics , Primary Cell Culture , Pyrimidines/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Th1 Cells/metabolism
7.
Mucosal Immunol ; 14(3): 679-690, 2021 05.
Article in English | MEDLINE | ID: mdl-33568785

ABSTRACT

Inflammatory bowel disease is characterized by an exacerbated intestinal immune response, but the critical mechanisms regulating immune activation remain incompletely understood. We previously reported that the TNF-superfamily molecule TNFSF14 (LIGHT) is required for preventing severe disease in mouse models of colitis. In addition, deletion of lymphotoxin beta receptor (LTßR), which binds LIGHT, also led to aggravated colitis pathogenesis. Here, we aimed to determine the cell type(s) requiring LTßR and the mechanism critical for exacerbation of colitis. Specific deletion of LTßR in neutrophils (LTßRΔN), but not in several other cell types, was sufficient to induce aggravated colitis and colonic neutrophil accumulation. Mechanistically, RNA-Seq analysis revealed LIGHT-induced suppression of cellular metabolism, and mitochondrial function, that was dependent on LTßR. Functional studies confirmed increased mitochondrial mass and activity, associated with excessive mitochondrial ROS production and elevated glycolysis at steady-state and during colitis. Targeting these metabolic changes rescued exacerbated disease severity. Our results demonstrate that LIGHT signals to LTßR on neutrophils to suppress metabolic activation and thereby prevents exacerbated immune pathogenesis during colitis.


Subject(s)
Colitis/immunology , Inflammatory Bowel Diseases/immunology , Lymphotoxin beta Receptor/metabolism , Mitochondria/metabolism , Neutrophils/metabolism , Activation, Metabolic , Animals , Dextran Sulfate , Disease Models, Animal , Disease Progression , Humans , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
8.
Nat Metab ; 1(1): 86-97, 2019 01.
Article in English | MEDLINE | ID: mdl-31528845

ABSTRACT

Decreased adipose tissue oxygen tension and increased HIF-1α expression can trigger adipose tissue inflammation and dysfunction in obesity. Our current understanding of obesity-associated decreased adipose tissue oxygen tension is mainly focused on changes in oxygen supply and angiogenesis. Here, we demonstrate that increased adipocyte O2 demand, mediated by ANT2 activity, is the dominant cause of adipocyte hypoxia. Deletion of adipocyte Ant2 improves obesity-induced intracellular adipocyte hypoxia by decreasing obesity-induced adipocyte oxygen demand, without effects on mitochondrial number or mass, or oligomycin-sensitive respiration. This led to decreased adipose tissue HIF-1α expression and inflammation with improved glucose tolerance and insulin resistance in both a preventative or therapeutic setting. Our results suggest that ANT2 may be a target for the development of insulin sensitizing drugs and that ANT2 inhibition might have clinical utility.


Subject(s)
Adenine Nucleotide Translocator 2/deficiency , Adipocytes/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Insulin Resistance/genetics , Obesity/etiology , Obesity/metabolism , Adipose Tissue/metabolism , Animals , Apoptosis , Fibrosis , Gene Expression Regulation , Gene Knockdown Techniques , Glucose/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/etiology , Inflammation/metabolism , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Oxygen/metabolism
9.
Cell Metab ; 28(3): 490-503.e7, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30043752

ABSTRACT

Long-chain fatty acid (LCFA) oxidation has been shown to play an important role in interleukin-4 (IL-4)-mediated macrophage polarization (M(IL-4)). However, many of these conclusions are based on the inhibition of carnitine palmitoyltransferase-1 with high concentrations of etomoxir that far exceed what is required to inhibit enzyme activity (EC90 < 3 µM). We employ genetic and pharmacologic models to demonstrate that LCFA oxidation is largely dispensable for IL-4-driven polarization. Unexpectedly, high concentrations of etomoxir retained the ability to disrupt M(IL-4) polarization in the absence of Cpt1a or Cpt2 expression. Although excess etomoxir inhibits the adenine nucleotide translocase, oxidative phosphorylation is surprisingly dispensable for M(IL-4). Instead, the block in polarization was traced to depletion of intracellular free coenzyme A (CoA), likely resulting from conversion of the pro-drug etomoxir into active etomoxiryl CoA. These studies help explain the effect(s) of excess etomoxir on immune cells and reveal an unappreciated role for CoA metabolism in macrophage polarization.


Subject(s)
Acyl Coenzyme A/physiology , Enzyme Inhibitors/pharmacology , Epoxy Compounds/pharmacology , Homeostasis/drug effects , Macrophages , Mitochondria , 3T3 Cells , A549 Cells , Animals , Carnitine O-Palmitoyltransferase/metabolism , Fatty Acids/metabolism , HCT116 Cells , Hep G2 Cells , Humans , Interleukin-4/metabolism , Liver/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Oxidative Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
10.
Neurochem Int ; 117: 15-22, 2018 07.
Article in English | MEDLINE | ID: mdl-29042253

ABSTRACT

At abnormally elevated levels of intracellular Ca2+, mitochondrial Ca2+ uptake may compromise mitochondrial electron transport activities and trigger membrane permeability changes that allow for release of cytochrome c and other mitochondrial apoptotic proteins into the cytosol. In this study, a clinically relevant canine cardiac arrest model was used to assess the effects of global cerebral ischemia and reperfusion on mitochondrial Ca2+ uptake capacity, Ca2+ uptake-mediated inhibition of respiration, and Ca2+-induced cytochrome c release, as measured in vitro in a K+-based medium in the presence of Mg2+, ATP, and NADH-linked oxidizable substrates. Maximum Ca2+ uptake by frontal cortex mitochondria was significantly lower following 10 min cardiac arrest compared to non-ischemic controls. Mitochondria from ischemic brains were also more sensitive to the respiratory inhibition associated with accumulation of large levels of Ca2+. Cytochrome c was released from brain mitochondria in vitro in a Ca2+-dose-dependent manner and was more pronounced following both 10 min of ischemia alone and following 24 h reperfusion, in comparison to mitochondria from non-ischemic Shams. These effects of ischemia and reperfusion on brain mitochondria could compromise intracellular Ca2+ homeostasis, decrease aerobic and increase anaerobic cerebral energy metabolism, and potentiate the cytochrome c-dependent induction of apoptosis, when re-oxygenated mitochondria are exposed to abnormally high levels of intracellular Ca2+.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Calcium/metabolism , Cytochromes c/metabolism , Mitochondria/metabolism , Animals , Brain/pathology , Brain Ischemia/pathology , Dogs , Female , Heart Arrest/metabolism , Heart Arrest/pathology , Mitochondria/pathology
11.
Anal Biochem ; 552: 60-65, 2018 07 01.
Article in English | MEDLINE | ID: mdl-28987935

ABSTRACT

Activities of enzymes localized to the mitochondrial matrix of mammalian cells are often critical regulatory steps in cellular metabolism. As such, measurement of matrix enzyme activities in response to genetic modifications or drug interventions is often desired. However, measurements in intact cells are often hampered by the presence of other isozymes in the cytoplasm as well as the inability to deliver enzyme substrates across cellular membranes. Classic approaches to liberate matrix enzymes utilize harsh treatments that disrupt intracellular architecture or require significant starting material to allow mitochondrial isolation prior to sample extraction. We describe a method using permeabilization reagents for both the plasma and mitochondrial membranes to allow in situ measurement of matrix enzyme activities. It is applied to adherent cell monolayers in 96-well plates treated with perfringolysin O to permeabilize the plasma membrane and alamethicin to permeabilize the mitochondrial inner membrane. We present three examples validated with inhibitor sensitivity: (i) Complex I-mediated oxygen consumption driven by NADH, (ii) ATP hydrolysis by the F1FO complex measuring pH changes in an Agilent Seahorse XF Analyzer, and (iii) Mitochondrial glutaminase (GLS1) activity in a coupled reaction monitoring NADH fluorescence in a plate reader.


Subject(s)
Bacterial Toxins/pharmacology , Cell Membrane Permeability/drug effects , Hemolysin Proteins/pharmacology , Mitochondrial Membranes/drug effects , A549 Cells , Glutaminase/metabolism , Hep G2 Cells , Humans , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , NAD/metabolism , Oxygen Consumption
12.
J Cell Biol ; 216(4): 1091-1105, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28254829

ABSTRACT

Glutamate is the dominant excitatory neurotransmitter in the brain, but under conditions of metabolic stress it can accumulate to excitotoxic levels. Although pharmacologic modulation of excitatory amino acid receptors is well studied, minimal consideration has been given to targeting mitochondrial glutamate metabolism to control neurotransmitter levels. Here we demonstrate that chemical inhibition of the mitochondrial pyruvate carrier (MPC) protects primary cortical neurons from excitotoxic death. Reductions in mitochondrial pyruvate uptake do not compromise cellular energy metabolism, suggesting neuronal metabolic flexibility. Rather, MPC inhibition rewires mitochondrial substrate metabolism to preferentially increase reliance on glutamate to fuel energetics and anaplerosis. Mobilizing the neuronal glutamate pool for oxidation decreases the quantity of glutamate released upon depolarization and, in turn, limits the positive-feedback cascade of excitotoxic neuronal injury. The finding links mitochondrial pyruvate metabolism to glutamatergic neurotransmission and establishes the MPC as a therapeutic target to treat neurodegenerative diseases characterized by excitotoxicity.


Subject(s)
Cell Death/physiology , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Neurons/metabolism , Neurons/physiology , Pyruvic Acid/metabolism , Animals , Energy Metabolism/physiology , Glutamic Acid/metabolism , Mitochondrial Proteins , Monocarboxylic Acid Transporters , Neurodegenerative Diseases/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Solute Carrier Proteins
13.
Free Radic Biol Med ; 82: 63-72, 2015 May.
Article in English | MEDLINE | ID: mdl-25578654

ABSTRACT

Polyunsaturated fatty acid (PUFA) peroxidation is initiated by hydrogen atom abstraction at bis-allylic sites and sets in motion a chain reaction that generates multiple toxic products associated with numerous disorders. Replacement of bis-allylic hydrogens of PUFAs with deuterium atoms (D-PUFAs), termed site-specific isotope reinforcement, inhibits PUFA peroxidation and confers cell protection against oxidative stress. We demonstrate that structurally diverse deuterated PUFAs similarly protect against oxidative stress-induced injury in both yeast and mammalian (myoblast H9C2) cells. Cell protection occurs specifically at the lipid peroxidation step, as the formation of isoprostanes, immediate products of lipid peroxidation, is drastically suppressed by D-PUFAs. Mitochondrial bioenergetics function is a likely downstream target of oxidative stress and a subject of protection by D-PUFAs. Pretreatment of cells with D-PUFAs is shown to prevent inhibition of maximal uncoupler-stimulated respiration as well as increased mitochondrial uncoupling, in response to oxidative stress induced by agents with diverse mechanisms of action, including t-butylhydroperoxide, ethacrynic acid, or ferrous iron. Analysis of structure-activity relationships of PUFAs harboring deuterium at distinct sites suggests that there may be a mechanism supplementary to the kinetic isotope effect of deuterium abstraction off the bis-allylic sites that accounts for the protection rendered by deuteration of PUFAs. Paradoxically, PUFAs with partially deuterated bis-allylic positions that retain vulnerable hydrogen atoms (e.g., monodeuterated 11-D1-Lin) protect in a manner similar to that of PUFAs with completely deuterated bis-allylic positions (e.g., 11,11-D2-Lin). Moreover, inclusion of just a fraction of deuterated PUFAs (20-50%) in the total pool of PUFAs preserves mitochondrial respiratory function and confers cell protection. The results indicate that the therapeutic potential of D-PUFAs may derive from the preservation of mitochondrial function.


Subject(s)
Antioxidants/pharmacology , Fatty Acids, Unsaturated/pharmacology , Mitochondria/metabolism , Oxidative Stress/drug effects , Saccharomyces cerevisiae/metabolism , Animals , Cell Line , Cell Respiration , Deuterium , Energy Metabolism , Ethacrynic Acid/pharmacology , Lipid Peroxidation/physiology , Rats , Structure-Activity Relationship , tert-Butylhydroperoxide/pharmacology
14.
J Bioenerg Biomembr ; 46(6): 471-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25248416

ABSTRACT

Mitochondrial reactive oxygen species (ROS) metabolism is unique in that mitochondria both generate and scavenge ROS. Recent estimates of ROS scavenging capacity of brain mitochondria are surprisingly high, ca. 9-12 nmol H2O2/min/mg, which is ~100 times higher than the rate of ROS generation. This raises a question whether brain mitochondria are a source or a sink of ROS. We studied the interaction between ROS generation and scavenging in mouse brain mitochondria by measuring the rate of removal of H2O2 added at a concentration of 0.4 µM, which is close to the reported physiological H2O2 concentrations in tissues, under conditions of low and high levels of mitochondrial H2O2 generation. With NAD-linked substrates, the rate of H2O2 generation by mitochondria was ~50-70 pmol/min/mg. The H2O2 scavenging dynamics was best approximated by the first order reaction equation. H2O2 scavenging was not affected by the uncoupling of mitochondria, phosphorylation of added ADP, or the genetic ablation of glutathione peroxidase 1, but decreased in the absence of respiratory substrates, in the presence of thioredoxin reductase inhibitor auranofin, or in partially disrupted mitochondria. With succinate, the rate of H2O2 generation was ~2,200-2,900 pmol/min/mg; the scavenging of added H2O2 was masked by a significant accumulation of generated H2O2 in the assay medium. The obtained data were fitted into a simple model that reasonably well described the interaction between H2O2 scavenging and production. It showed that mitochondria are neither a sink nor a source of H2O2, but can function as both at the same time, efficiently stabilizing exogenous H2O2 concentration at a level directly proportional to the ratio of the H2O2 generation rate to the rate constant of the first order scavenging reaction.


Subject(s)
Brain/metabolism , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Animals , Mice , Oxidative Stress , Reactive Oxygen Species
15.
EMBO Mol Med ; 5(6): 904-18, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23703906

ABSTRACT

Miner1 is a redox-active 2Fe2S cluster protein. Mutations in Miner1 result in Wolfram Syndrome, a metabolic disease associated with diabetes, blindness, deafness, and a shortened lifespan. Embryonic fibroblasts from Miner1(-/-) mice displayed ER stress and showed hallmarks of the unfolded protein response. In addition, loss of Miner1 caused a depletion of ER Ca(2+) stores, a dramatic increase in mitochondrial Ca(2+) load, increased reactive oxygen and nitrogen species, an increase in the GSSG/GSH and NAD(+)/NADH ratios, and an increase in the ADP/ATP ratio consistent with enhanced ATP utilization. Furthermore, mitochondria in fibroblasts lacking Miner1 displayed ultrastructural alterations, such as increased cristae density and punctate morphology, and an increase in O2 consumption. Treatment with the sulphydryl anti-oxidant N-acetylcysteine reversed the abnormalities in the Miner1 deficient cells, suggesting that sulphydryl reducing agents should be explored as a treatment for this rare genetic disease.


Subject(s)
Calcium/metabolism , Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Sulfhydryl Compounds/chemistry , Unfolded Protein Response , Adenosine Triphosphate/metabolism , Animals , Antioxidants/pharmacology , Autophagy-Related Proteins , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line , Glutathione/metabolism , Glutathione Disulfide/metabolism , Mice , Mitochondria/metabolism , Mitochondria/ultrastructure , NAD/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Oxidation-Reduction , Sulfhydryl Compounds/metabolism , Unfolded Protein Response/drug effects , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology
16.
Proc Natl Acad Sci U S A ; 110(14): 5422-7, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23513224

ABSTRACT

Facilitated pyruvate transport across the mitochondrial inner membrane is a critical step in carbohydrate, amino acid, and lipid metabolism. We report that clinically relevant concentrations of thiazolidinediones (TZDs), a widely used class of insulin sensitizers, acutely and specifically inhibit mitochondrial pyruvate carrier (MPC) activity in a variety of cell types. Respiratory inhibition was overcome with methyl pyruvate, localizing the effect to facilitated pyruvate transport, and knockdown of either paralog, MPC1 or MPC2, decreased the EC50 for respiratory inhibition by TZDs. Acute MPC inhibition significantly enhanced glucose uptake in human skeletal muscle myocytes after 2 h. These data (i) report that clinically used TZDs inhibit the MPC, (ii) validate that MPC1 and MPC2 are obligatory components of facilitated pyruvate transport in mammalian cells, (iii) indicate that the acute effect of TZDs may be related to insulin sensitization, and (iv) establish mitochondrial pyruvate uptake as a potential therapeutic target for diseases rooted in metabolic dysfunction.


Subject(s)
Cell Respiration/drug effects , Membrane Transport Proteins/metabolism , Metabolic Networks and Pathways/physiology , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Thiazolidinediones/pharmacology , Acrylates/pharmacology , Analysis of Variance , Animals , Anion Transport Proteins , Blotting, Western , Cell Line , Cytochromes c/metabolism , Glucose/metabolism , Humans , Membrane Potential, Mitochondrial/physiology , Mice , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/metabolism , Monocarboxylic Acid Transporters , Muscle, Skeletal/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Solute Carrier Proteins , Thiazolidinediones/metabolism
17.
PLoS Biol ; 10(9): e1001394, 2012.
Article in English | MEDLINE | ID: mdl-23049480

ABSTRACT

Bax/Bak-mediated mitochondrial outer membrane permeabilization (MOMP) is essential for "intrinsic" apoptotic cell death. Published studies used synthetic liposomes to reveal an intrinsic pore-forming activity of Bax, but it is unclear how other mitochondrial outer membrane (MOM) proteins might facilitate this function. We carefully analyzed the kinetics of Bax-mediated pore formation in isolated MOMs, with some unexpected results. Native MOMs were more sensitive than liposomes to added Bax, and MOMs displayed a lag phase not observed with liposomes. Heat-labile MOM proteins were required for this enhanced response. A two-tiered mathematical model closely fit the kinetic data: first, Bax activation promotes the assembly of a multimeric complex, which then catalyzes the second reaction, Bax-dependent pore formation. Bax insertion occurred immediately upon Bax addition, prior to the end of the lag phase. Permeabilization kinetics were affected in a reciprocal manner by [cBid] and [Bax], confirming the "hit-and-run" hypothesis of cBid-induced direct Bax activation. Surprisingly, MOMP rate constants were linearly related to [Bax], implying that Bax acts non-cooperatively. Thus, the oligomeric catalyst is distinct from Bax. Moreover, contrary to common assumption, pore formation kinetics depend on Bax monomers, not oligomers. Catalyst formation exhibited a sharp transition in activation energy at ∼28°C, suggesting a role for membrane lipid packing. Furthermore, catalyst formation was strongly inhibited by chemical antagonists of the yeast mitochondrial fission protein, Dnm1. However, the mammalian ortholog, Drp1, was undetectable in mitochondrial outer membranes. Moreover, ATP and GTP were dispensable for MOMP. Thus, the data argue that oligomerization of a catalyst protein, distinct from Bax and Drp1, facilitates MOMP, possibly through a membrane-remodeling event.


Subject(s)
Biocatalysis , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Protein Multimerization , bcl-2-Associated X Protein/metabolism , Animals , BH3 Interacting Domain Death Agonist Protein/metabolism , Biocatalysis/drug effects , Dynamin I/metabolism , Humans , Kinetics , Liposomes/metabolism , Male , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Permeability Transition Pore , Models, Biological , Permeability/drug effects , Protein Multimerization/drug effects , Quinazolinones/pharmacology , Rats , Rats, Sprague-Dawley , Thermodynamics
18.
Mol Cell Proteomics ; 11(7): M111.014746, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22361236

ABSTRACT

Eicosanoids constitute a diverse class of bioactive lipid mediators that are produced from arachidonic acid and play critical roles in cell signaling and inflammatory aspects of numerous diseases. We have previously quantified eicosanoid metabolite production in RAW264.7 macrophage cells in response to Toll-like receptor 4 signaling and analyzed the levels of transcripts coding for the enzymes involved in the eicosanoid metabolite biosynthetic pathways. We now report the quantification of changes in protein levels under similar experimental conditions in RAW264.7 macrophages by multiple reaction monitoring mass spectrometry, an accurate targeted protein quantification method. The data complete the first fully integrated genomic, proteomic, and metabolomic analysis of the eicosanoid biochemical pathway.


Subject(s)
Arachidonic Acid/metabolism , Biosynthetic Pathways/drug effects , Eicosanoids/biosynthesis , Inflammation/metabolism , Macrophages/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Line , Inflammation/chemically induced , Lipid Metabolism/drug effects , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mass Spectrometry , Metabolomics , Mice , Proteomics , Signal Transduction/drug effects
19.
J Lipid Res ; 51(9): 2785-97, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20574076

ABSTRACT

Lipids orchestrate biological processes by acting remotely as signaling molecules or locally as membrane components that modulate protein function. Detailed insight into lipid function requires knowledge of the subcellular localization of individual lipids. We report an analysis of the subcellular lipidome of the mammalian macrophage, a cell type that plays key roles in inflammation, immune responses, and phagocytosis. Nuclei, mitochondria, endoplasmic reticulum (ER), plasmalemma, and cytoplasm were isolated from RAW 264.7 macrophages in basal and activated states. Subsequent lipidomic analyses of major membrane lipid categories identified 229 individual/isobaric species, including 163 glycerophospholipids, 48 sphingolipids, 13 sterols, and 5 prenols. Major subcellular compartments exhibited substantially divergent glycerophospholipid profiles. Activation of macrophages by the Toll-like receptor 4-specific lipopolysaccharide Kdo(2)-lipid A caused significant remodeling of the subcellular lipidome. Some changes in lipid composition occurred in all compartments (e.g., increases in the levels of ceramides and the cholesterol precursors desmosterol and lanosterol). Other changes were manifest in specific organelles. For example, oxidized sterols increased and unsaturated cardiolipins decreased in mitochondria, whereas unsaturated ether-linked phosphatidylethanolamines decreased in the ER. We speculate that these changes may reflect mitochondrial oxidative stress and the release of arachidonic acid from the ER in response to cell activation.


Subject(s)
Lipids/analysis , Macrophages , Organelles/chemistry , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , Computational Biology/methods , Databases, Factual , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Macrophages/metabolism , Macrophages/ultrastructure , Mice , Oxidative Stress
20.
Mol Cell Proteomics ; 9(2): 388-402, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19884172

ABSTRACT

Compartmentalization of biological processes and the associated cellular components is crucial for cell function. Typically, the location of a component is revealed through a co-localization and/or co-purification with an organelle marker. Therefore, the identification of reliable markers is critical for a thorough understanding of cellular function and dysfunction. We fractionated macrophage-like RAW264.7 cells, both in the resting and endotoxin-activated states, into six fractions representing the major organelles/compartments: nuclei, mitochondria, cytoplasm, endoplasmic reticulum, and plasma membrane as well as an additional dense microsomal fraction. The identity of the first five of these fractions was confirmed via the distribution of conventional enzymatic markers. Through a quantitative liquid chromatography/mass spectrometry-based proteomics analysis of the fractions, we identified 50-member ensembles of marker proteins ("marker ensembles") specific for each of the corresponding organelles/compartments. Our analysis attributed 206 of the 250 marker proteins ( approximately 82%) to organelles that are consistent with the location annotations in the public domain (obtained using DAVID 2008, EntrezGene, Swiss-Prot, and references therein). Moreover, we were able to correct locations for a subset of the remaining proteins, thus proving the superior power of analysis using multiple organelles as compared with an analysis using one specific organelle. The marker ensembles were used to calculate the organelle composition of the six above mentioned subcellular fractions. Knowledge of the precise composition of these fractions can be used to calculate the levels of metabolites in the pure organelles. As a proof of principle, we applied these calculations to known mitochondria-specific lipids (cardiolipins and ubiquinones) and demonstrated their exclusive mitochondrial location. We speculate that the organelle-specific protein ensembles may be used to systematically redefine originally morphologically defined organelles as biochemical entities.


Subject(s)
Biomarkers/metabolism , Cell Fractionation/methods , Organelles/metabolism , Proteomics/methods , Animals , Cell Line , Chromatography, Liquid , Computational Biology , DNA/metabolism , Lipids/analysis , Mice , Microsomes/metabolism , Nanotechnology , Proliferating Cell Nuclear Antigen/metabolism , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...