Downregulation of adipose glutathione S-transferase A4 leads to increased protein carbonylation, oxidative stress, and mitochondrial dysfunction

Jessica M Curtis, Paul A Grimsrud, Wendy S Wright, Xin Xu, Rocio E Foncea, David W Graham, Jonathan R Brestoff, Brian M Wiczer, Olga Ilkayeva, Katherine Cianflone, Deborah E Muoio, Edgar A Arriaga, David A Bernlohr, Jessica M Curtis, Paul A Grimsrud, Wendy S Wright, Xin Xu, Rocio E Foncea, David W Graham, Jonathan R Brestoff, Brian M Wiczer, Olga Ilkayeva, Katherine Cianflone, Deborah E Muoio, Edgar A Arriaga, David A Bernlohr

Abstract

Objective: Peripheral insulin resistance is linked to an increase in reactive oxygen species (ROS), leading in part to the production of reactive lipid aldehydes that modify the side chains of protein amino acids in a reaction termed protein carbonylation. The primary enzymatic method for lipid aldehyde detoxification is via glutathione S-transferase A4 (GSTA4) dependent glutathionylation. The objective of this study was to evaluate the expression of GSTA4 and the role(s) of protein carbonylation in adipocyte function.

Research design and methods: GSTA4-silenced 3T3-L1 adipocytes and GSTA4-null mice were evaluated for metabolic processes, mitochondrial function, and reactive oxygen species production. GSTA4 expression in human obesity was evaluated using microarray analysis.

Results: GSTA4 expression is selectively downregulated in adipose tissue of obese insulin-resistant C57BL/6J mice and in human obesity-linked insulin resistance. Tumor necrosis factor-alpha treatment of 3T3-L1 adipocytes decreased GSTA4 expression, and silencing GSTA4 mRNA in cultured adipocytes resulted in increased protein carbonylation, increased mitochondrial ROS, dysfunctional state 3 respiration, and altered glucose transport and lipolysis. Mitochondrial function in adipocytes of lean or obese GSTA4-null mice was significantly compromised compared with wild-type controls and was accompanied by an increase in superoxide anion.

Conclusions: These results indicate that downregulation of GSTA4 in adipose tissue leads to increased protein carbonylation, ROS production, and mitochondrial dysfunction and may contribute to the development of insulin resistance and type 2 diabetes.

Figures

FIG. 1.
FIG. 1.
Expression of oxidative stress–responsive genes in adipose tissue in obesity. C57BL/6J mice were fed either a low-fat or high-fat diet for 9–12 weeks, and mRNA was isolated from the indicated tissue. A: Expression of the indicated genes in epididymal WAT analyzed by qPCR (lean, n = 8; obese, n = 7). NQO-1, NADPH quninone oxidoreductase 1. B: Expression of GST isozymes in WAT of lean and obese male and female mice (male, lean, n = 8; male, obese, n = 7; female, lean, n = 5; female, obese, n = 4). C: Expression of GSTA4 in various tissues or cells from lean and obese male mice (n = 8). D: Expression of GSTA4 in C57BL/6J mice (B6) relative to that in adipose tissue of ob/ob mice. TBP, TATA-box binding protein; TFIIE, transcription factor II E. *P < 0.05; **P < 0.01.
FIG. 2.
FIG. 2.
Effect of TNF-α treatment on GSTA4 expression in 3T3–L1 adipocytes. qPCR analysis of GSTA4 mRNA expression in day-8 3T3–L1 adipocytes normalized to TFIIE as a function of (A) TNF-α level after 24 h or (B) time of treatment with 1 nmol/l TNF-α. *P < 0.05; **P < 0.01 relative to control samples.
FIG. 3.
FIG. 3.
Expression of human GSTA4 in obesity and insulin resistance. A: Relative expression of GSTA4 in omental and subcutaneous white adipose tissues of patients characterized as lean insulin sensitive (Lean), obese insulin sensitive (OB-IS), or obese insulin resistant (OB-IR). Data are expressed using means ± SEM. B: Correlation of GSTA4 mRNA expression in omental or subcutaneous adipose with patient BMI in kg/m2. C: Correlation of GSTA4 mRNA expression in omental or subcutaneous adipose with HOMA-IR, calculated as [insulin (μU/ml) × glucose (mmol/l)]/22.5. Each data point represents one individual.
FIG. 4.
FIG. 4.
GSTA4 silencing and protein carbonylation in 3T3–L1 adipocytes. A: Relative levels of GSTA4 and GSTA3 mRNA were quantified by qPCR (n = 6 per group). GSTA4 expression was normalized to TFIIE, and GSTA3 expression was normalized to 36B4. B: Expression of adipogenic marker proteins (n = 6 per group). C: Protein carbonylation in GSTA4 knockdown and Scr control cells. Protein bands found to have increased carbonylation in the GSTA4 knockdown adipocytes are indicated (*). The ∼145-kDa band (**) was digested and subjected to liquid chromatography–electrospray ionization tandem mass spectrometry for protein identification detailed in supplementary Table 2. S, scrambled, −BH, minus biotin hydrazide. C: A composite in which the −BH lane has been moved from the same digital image (at the same exposure) to be adjacent to the experimental lanes.
FIG. 5.
FIG. 5.
Glucose and lipid metabolism in GSTA4 knockdown and Scr adipocytes. A: Transport of 2-deoxyglucose (2-DG) in GSTA4 knockdown and Scr adipocytes under basal (left) and 100 nmol/l insulin-stimulated (right) conditions. B: Fold stimulation of hexose transport in GSTA4 knockdown and Scr 3T3–L1 adipocytes (n = 9). C: Expression of glucose transporters GLUT1 and GLUT4 (n = 6 per group). D: NAD+, NADH, and NAD+/NADH in day 7 GSTA4 knockdown and Scr adipocytes (n = 6). E: L(+)-lactate in the medium of GSTA4 knockdown and Scr cells (n = 6). F: Organic acids from GSTA4 knockdown and Scr adipocyte cell lysates. Basal (G) and forskolin-stimulated (H) lipolysis in Scr and GSTA4 knockdown 3T3–L1 adipocytes. I: β-oxidation of [14C]-palmitate in Scr and knockdown adipocytes (n = 6). G and H: Analyzed by two-way ANOVA with Bonferroni post hoc analysis. *P < 0.05; **P < 0.01.
FIG. 6.
FIG. 6.
Mitochondria function in GSTA4 knockdown and Scr 3T3–L1 adipocytes. Mitochondrial oxygen consumption in Scr (A) or GSTA4 knockdown (B) adipocytes. C: Oxygen consumption rates for Scr and GSTA4 knockdown adipocytes (n = 3). Statistics calculated by two-way analysis with Bonferroni post hoc analysis. D: Mitochondrial matrix superoxide production in GSTA4 knockdown and Scr adipocytes (n = 3).
FIG. 7.
FIG. 7.
Expression of genes and proteins linked to mitochondrial biogenesis. A: Expression of transcription factors and target mRNA in GSTA4-silenced (open bars) and scrambled (closed bars) adipocytes (n = 6). B: Mitochondrial protein expression in GSTA4 knockdown and Scr adipocytes (n = 3–6). C: Expression of COX II and cytochrome b DNA relative to UCP2 DNA in GSTA4 knockdown and Scr adipocytes (n = 6). D: Activity of ATP synthase in GSTA4-silenced and scrambled adipocytes and level of ATP synthase α-subunit protein (n = 3). *P < 0.05; **P < 0.01.
FIG. 8.
FIG. 8.
Mitochondrial function and expression in adipose tissue from C57BL/6J and GSTA4-null (−/−) mice. Mitochondria were isolated from EWAT of 4- to 5-month-old mice maintained on a standard chow (lean) or high-fat (obese) diets. A: Mitochondrial oxygen consumption in lean and obese wild-type (+/+) and GSTA4-null (−/−) adipose. The mitochondrial protein concentrations were 0.22 (lean +/+), 0.23 (obese +/+), 0.20 (lean −/−), and 0.23 (obese −/−) mg/ml. B: Oxygen consumption rates for all four groups (n = 3). Statistics calculated using two-way ANOVA for each state with Holm-Sidak post hoc analysis. Variation due to loss of GSTA4 in state 2: P = 0.1; variation due to obesity in state 3: P = 0.03. C: Mitochondrial matrix superoxide production (n = 3). Statistics calculated using two-way ANOVA with Bonferroni post hoc analysis. Effect of obesity: P = 0.0009; effect of GSTA4: P = 0.0018. D: Quantitation of mitochondrial protein carbonylation in EWAT from lean wild-type and GSTA4-null mice (n = 6). *P < 0.05, **P < 0.01, ***P < 0.001.

References

    1. Ogden CL, Carroll MD, Curtin LR, McDowell MA, Tabak CJ, Flegal KM. Prevalence of overweight and obesity in the United States, 1999–2004. JAMA 2006;295:1549–1555
    1. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest 2004;114:1752–1761
    1. Talior I, Yarkoni M, Bashan N, Eldar-Finkelman H. Increased glucose uptake promotes oxidative stress and PKC-delta activation in adipocytes of obese, insulin-resistant mice. Am J Physiol Endocrinol Metab 2003;285:E295–E302
    1. Anderson EJ, Lustig ME, Boyle KE, Woodlief TL, Kane DA, Lin CT, Price JW, III, Kang L, Rabinovitch PS, Szeto HH, Houmard JA, Cortright RN, Wasserman DH, Neufer PD. Mitochondrial H2O2 emission and cellular redox state link excess fat intake to insulin resistance in both rodents and humans. J Clin Invest 2009February2 pii: 37048 doi: . [Epub ahead of print]
    1. Houstis N, Rosen ED, Lander ES. Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature 2006;440:944–948
    1. Lin Y, Berg AH, Iyengar P, Lam TK, Giacca A, Combs TP, Rajala MW, Du X, Rollman B, Li W, Hawkins M, Barzilai N, Rhodes CJ, Fantus IG, Brownlee M, Scherer PE. The hyperglycemia-induced inflammatory response in adipocytes: the role of reactive oxygen species. J Biol Chem 2005;280:4617–4626
    1. Soares AF, Guichardant M, Cozzone D, Bernoud-Hubac N, Bouzaïdi-Tiali N, Lagarde M, Géloën A. Effects of oxidative stress on adiponectin secretion and lactate production in 3T3–L1 adipocytes. Free Radic Biol Med 2005;38:882–889
    1. Bays H, Mandarino L, DeFronzo RA. Role of the adipocyte, free fatty acids, and ectopic fat in pathogenesis of type 2 diabetes mellitus: peroxisomal proliferator-activated receptor agonists provide a rational therapeutic approach. J Clin Endocrinol Metab 2004;89:463–478
    1. Zarković N, Zarković K, Schaur RJ, Stolc S, Schlag G, Redl H, Waeg G, Borović S, Loncarić I, Jurić G, Hlavka V. 4-Hydroxynonenal as a second messenger of free radicals and growth modifying factor. Life Sci 1999;65:1901–1904
    1. Sayre LM, Lin D, Yuan Q, Zhu X, Tang X. Protein adducts generated from products of lipid oxidation: focus on HNE and one. Drug Metab Rev 2006;38:651–675
    1. Awasthi YC, Sharma R, Cheng JZ, Yang Y, Sharma A, Singhal SS, Awasthi S. Role of 4-hydroxynonenal in stress-mediated apoptosis signaling. Mol Aspects Med 2003;24:219–230
    1. Ueda K, Ueyama T, Yoshida K, Kimura H, Ito T, Shimizu Y, Oka M, Tsuruo Y, Ichinose M. Adaptive HNE-Nrf2-HO-1 pathway against oxidative stress is associated with acute gastric mucosal lesions. Am J Physiol Gastrointest Liver Physiol 2008;295:G460–G469
    1. Piantadosi CA, Suliman HB. Mitochondrial transcription factor A induction by redox activation of nuclear respiratory factor 1. J Biol Chem 2006;281:324–333
    1. Grimsrud PA, Xie H, Griffin TJ, Bernlohr DA. Oxidative stress and covalent modification of protein with bioactive aldehydes. J Biol Chem 2008;283:21837–21841
    1. Grimsrud PA, Picklo MJ, Sr, Griffin TJ, Bernlohr DA. Carbonylation of adipose proteins in obesity and insulin resistance: identification of adipocyte fatty acid-binding protein as a cellular target of 4-hydroxynonenal. Mol Cell Proteomics 2007;6:624–637
    1. He NG, Singhal SS, Srivastava SK, Zimniak P, Awasthi YC, Awasthi S. Transfection of a 4-hydroxynonenal metabolizing glutathione S-transferase isozyme, mouse GSTA4–4, confers doxorubicin resistance to Chinese hamster ovary cells. Arch Biochem Biophys 1996;333:214–220
    1. Surwit RS, Kuhn CM, Cochrane C, McCubbin JA, Feinglos MN. Diet-induced type II diabetes in C57BL/6J mice. Diabetes 1988;37:1163–1167
    1. Malide D, Ramm G, Cushman SW, Slot JW. Immunoelectron microscopic evidence that GLUT4 translocation explains the stimulation of glucose transport in isolated rat white adipose cells. J Cell Sci 2000;113(Pt. 23):4203–4210
    1. Lobo S, Wiczer BM, Smith AJ, Hall AM, Bernlohr DA. Fatty acid metabolism in adipocytes: functional analysis of fatty acid transport proteins 1 and 4. J Lipid Res 2007;48:609–620
    1. Mogensen M, Sahlin K, Fernström M, Glintborg D, Vind BF, Beck-Nielsen H, Hj̸lund K. Mitochondrial respiration is decreased in skeletal muscle of patients with type 2 diabetes. Diabetes 2007;56:1592–1599
    1. Xu X, Arriaga EA. Qualitative determination of superoxide release at both sides of the mitochondrial inner membrane by capillary electrophoretic analysis of the oxidation products of triphenylphosphonium hydroethidine. Free Radic Biol Med 2009;46:905–913
    1. MacLaren R, Cui W, Simard S, Cianflone K. Influence of obesity and insulin sensitivity on insulin signaling genes in human omental and subcutaneous adipose tissue. J Lipid Res 2008;49:308–323
    1. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A 2001;98:5116–5121
    1. Moraes RC, Blondet A, Birkenkamp-Demtroeder K, Tirard J, Orntoft TF, Gertler A, Durand P, Naville D, Bégeot M. Study of the alteration of gene expression in adipose tissue of diet-induced obese mice by microarray and reverse transcription-polymerase chain reaction analyses. Endocrinology 2003;144:4773–4782
    1. Keller MP, Choi Y, Wang P, Davis DB, Rabaglia ME, Oler AT, Stapleton DS, Argmann C, Schueler KL, Edwards S, Steinberg HA, Chaibub Neto E, Kleinhanz R, Turner S, Hellerstein MK, Schadt EE, Yandell BS, Kendziorski C, Attie AD. A gene expression network model of type 2 diabetes links cell cycle regulation in islets with diabetes susceptibility. Genome Res 2008;18:706–716
    1. Hubatsch I, Ridderstrom M, Mannervik B. Human glutathione transferase A4–4: an alpha class enzyme with high catalytic efficiency in the conjugation of 4-hydroxynonenal and other genotoxic products of lipid peroxidation. Biochem J 1998;330(Pt. 1):175–179
    1. Humphries KM, Szweda LI. Selective inactivation of alpha-ketoglutarate dehydrogenase and pyruvate dehydrogenase: reaction of lipoic acid with 4-hydroxy-2-nonenal. Biochemistry 1998;37:15835–15841
    1. Yoo BS, Regnier FE. Proteomic analysis of carbonylated proteins in two-dimensional gel electrophoresis using avidin-fluorescein affinity staining. Electrophoresis 2004;25:1334–1341
    1. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O, Bain J, Stevens R, Dyck JR, Newgard CB, Lopaschuk GD, Muoio DM. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab 2008;7:45–56
    1. Koves TR, Noland RC, Bates AL, Henes ST, Muoio DM, Cortright RN. Subsarcolemmal and intermyofibrillar mitochondria play distinct roles in regulating skeletal muscle fatty acid metabolism. Am J Physiol Cell Physiol 2005;288:C1074–C1082
    1. Ritov VB, Menshikova EV, He J, Ferrell RE, Goodpaster BH, Kelley DE. Deficiency of subsarcolemmal mitochondria in obesity and type 2 diabetes. Diabetes 2005;54:8–14
    1. Valerio A, Cardile A, Cozzi V, Bracale R, Tedesco L, Pisconti A, Palomba L, Cantoni O, Clementi E, Moncada S, Carruba MO, Nisoli E. TNF-alpha downregulates eNOS expression and mitochondrial biogenesis in fat and muscle of obese rodents. J Clin Invest 2006;116:2791–2798
    1. Meany DL, Poe BG, Navratil M, Moraes CT, Arriaga EA. Superoxide released into the mitochondrial matrix. Free Radic Biol Med 2006;41:950–959
    1. Meany DL, Xie H, Thompson LV, Arriaga EA, Griffin TJ. Identification of carbonylated proteins from enriched rat skeletal muscle mitochondria using affinity chromatography-stable isotope labeling and tandem mass spectrometry. Proteomics 2007;7:1150–1163
    1. Engle MR, Singh SP, Czernik PJ, Gaddy D, Montague DC, Ceci JD, Yang Y, Awasthi S, Awasthi YC, Zimniak P. Physiological role of mGSTA4–4, a glutathione S-transferase metabolizing 4-hydroxynonenal: generation and analysis of mGsta4 null mouse. Toxicol Appl Pharmacol 2004;194:296–308
    1. Gallou-Kabani C, Vige A, Gross MS, Rabes JP, Boileau C, Larue-Achagiotis C, Tome D, Jais JP, Junien C. C57BL/6J and A/J mice fed a high-fat diet delineate components of metabolic syndrome. Obesity (Silver Spring) 2007;15:1996–2005
    1. Rasmussen JT, Rasmussen MS, Petersen TE. Cysteines involved in the interconversion between dehydrogenase and oxidase forms of bovine xanthine oxidoreductase. J Dairy Sci 2000;83:499–506
    1. Saksela M, Lapatto R, Raivio KO. Irreversible conversion of xanthine dehydrogenase into xanthine oxidase by a mitochondrial protease. FEBS Lett 1999;443:117–120
    1. Gaster M. Insulin resistance and the mitochondrial link: lessons from cultured human myotubes. Biochim Biophys Acta 2007;1772:755–765
    1. Jackson S, Bagstaff SM, Lynn S, Yeaman SJ, Turnbull DM, Walker M. Decreased insulin responsiveness of glucose uptake in cultured human skeletal muscle cells from insulin-resistant nondiabetic relatives of type 2 diabetic families. Diabetes 2000;49:1169–1177
    1. Hamrahian AH, Zhang JZ, Elkhairi FS, Prasad R, Ismail-Beigi F. Activation of Glut1 glucose transporter in response to inhibition of oxidative phosphorylation. Arch Biochem Biophys 1999;368:375–379
    1. Arner P. Control of lipolysis and its relevance to development of obesity in man. Diabetes Metab Rev 1988;4:507–515
    1. Ishii T, Itoh K, Ruiz E, Leake DS, Unoki H, Yamamoto M, Mann GE. Role of Nrf2 in the regulation of CD36 and stress protein expression in murine macrophages: activation by oxidatively modified LDL and 4-hydroxynonenal. Circ Res 2004;94:609–616
    1. Humphries KM, Yoo Y, Szweda LI. Inhibition of NADH-linked mitochondrial respiration by 4-hydroxy-2-nonenal. Biochemistry 1998;37:552–557
    1. Echtay KS, Esteves TC, Pakay JL, Jekabsons MB, Lambert AJ, Portero-Otín M, Pamplona R, Vidal-Puig AJ, Wang S, Roebuck SJ, Brand MD. A signalling role for 4-hydroxy-2-nonenal in regulation of mitochondrial uncoupling. EMBO J 2003;22:4103–4110
    1. Vieira HL, Belzacq AS, Haouzi D, Bernassola F, Cohen I, Jacotot E, Ferri KF, El Hamel C, Bartle LM, Melino G, Brenner C, Goldmacher V, Kroemer G. The adenine nucleotide translocator: a target of nitric oxide, peroxynitrite, and 4-hydroxynonenal. Oncogene 2001;20:4305–4316
    1. Tedesco L, Valerio A, Cervino C, Cardile A, Pagano C, Vettor R, Pasquali R, Carruba MO, Marsicano G, Lutz B, Pagotto U, Nisoli E. Cannabinoid type 1 receptor blockade promotes mitochondrial biogenesis through endothelial nitric oxide synthase expression in white adipocytes. Diabetes 2008;57:2028–2036

Source: PubMed

3
Subskrybuj