Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2744 2023-09-11 06:12:25 |
2 layout Meta information modification 2744 2023-09-11 07:20:19 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zhu, M.; Dagah, O.M.A.; Silaa, B.B.; Lu, J. Disruption of Thiol Homeostasis in NAFLD. Encyclopedia. Available online: https://encyclopedia.pub/entry/49003 (accessed on 07 September 2024).
Zhu M, Dagah OMA, Silaa BB, Lu J. Disruption of Thiol Homeostasis in NAFLD. Encyclopedia. Available at: https://encyclopedia.pub/entry/49003. Accessed September 07, 2024.
Zhu, Minghui, Omer M. A. Dagah, Billton Bryson Silaa, Jun Lu. "Disruption of Thiol Homeostasis in NAFLD" Encyclopedia, https://encyclopedia.pub/entry/49003 (accessed September 07, 2024).
Zhu, M., Dagah, O.M.A., Silaa, B.B., & Lu, J. (2023, September 11). Disruption of Thiol Homeostasis in NAFLD. In Encyclopedia. https://encyclopedia.pub/entry/49003
Zhu, Minghui, et al. "Disruption of Thiol Homeostasis in NAFLD." Encyclopedia. Web. 11 September, 2023.
Disruption of Thiol Homeostasis in NAFLD
Edit

Non-alcoholic fatty liver disease (NAFLD) is a clinical–pathological syndrome characterized by excessive deposition of fat and fatty degeneration in liver cells in the absence of excessive alcohol consumption or other known causes of liver disease.  The formation of disulfide bonds is a reversible process that can be achieved through thiol–disulfide interchange.

thioredoxin reactive oxygen species NAFLD gut microbiota dysbiosis

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is a clinical–pathological syndrome characterized by excessive deposition of fat and fatty degeneration in liver cells in the absence of excessive alcohol consumption or other known causes of liver disease. Pathologically, NAFLD is generally categorized into two types: simple fatty liver and non-alcoholic steatohepatitis (NASH) and can progress to fatty liver fibrosis and even liver cancer. Besides the direct harm from liver disease, NAFLD patients are at significantly increased risk of developing cardiovascular diseases, type 2 diabetes, and chronic renal insufficiency [1][2]. However, the specific pathogenesis of NAFLD/NASH remains unclear, and there is currently no specific treatment available. Clinical pharmacological research on NAFLD is actively underway.
NAFLD is a disease closely related to various factors, including genetics, environment, and diet. To emphasize its metabolic nature, in March 2020, an international expert group proposed renaming it as Metabolic Associated Fatty Liver Disease (MAFLD) [3]. When NAFLD progresses to NASH, the risk of liver fibrosis and liver cancer increases significantly. Consequently, in the natural development of NAFLD, the progression of the disease can be effectively stopped by improving NASH. The exact cause of NAFLD/NASH is still unknown, and the “two-hit” hypothesis proposes that insulin resistance in peripheral adipose tissue results in the breakdown of fats, elevated levels of free fatty acids in the bloodstream, and excessive transportation of fatty acids to the liver, ultimately leading to hepatic steatosis [4][5]. This is the “first hit”. Prolonged and excessive fat accumulation in the liver cannot be eliminated, causing mitochondrial dysfunction. This dysfunction leads to endoplasmic reticulum stress, oxidative stress, and the release of inflammatory factors, which exacerbate hepatocellular damage and contribute to the development of NASH from simple fatty liver disease, referred to as the “second hit”. Reduced secretion of adiponectin (ADPN) and elevated pro-inflammatory factors in peripheral adipose tissue further promote inflammatory responses and worsen insulin resistance, creating a vicious cycle [4]. Additionally, liver cell apoptosis is critical for the development of NASH. The pathogenesis of NAFLD/NASH is highly complex, and the current understanding has shifted from the “two-hit” to the “multiple parallel hits” theory, involving diverse factors such as oxidative stress, gut microbiota, insulin resistance, adipokines, genetics, and epigenetics, and others [5][6].

2. Disruption of Thiol Homeostasis in NAFLD

2.1. Sites of ROS Production

Reactive oxygen species (ROS) are inevitable byproducts of aerobic metabolism in living organisms, including superoxide (O2), hydrogen peroxide (H2O2), hydroxyl radicals (HO·), and so on. Among them, hydroxyl radicals are the most reactive, capable of attacking almost all biological molecules, inducing lipid peroxidation, and causing DNA double-strand breaks, making them the most destructive type of ROS [7][8], Superoxide (O2) serves as the precursor of other ROS. When O2 leaks from the mitochondrial respiratory chain, an O2 molecule accepting an electron generates O2, which can be dismutated by superoxide dismutase (SOD) into H2O2 and O2. H2O2 is not a free radical molecule but can be catalyzed to generate highly active HO· through the Fenton reaction when accepting an electron. The majority of ROS in cells originate from this process [9][10]. The excess ROS causes damage to mitochondrial DNA (mtDNA), respiratory chain complexes, lipids, and other components [11], which, in turn, further induces ROS production, creating a vicious cycle [12][13].
The other source sites of ROS include the endoplasmic reticulum (ER), a crucial site for protein synthesis, post-translational modification, processing, and folding in cells. In the ER lumen, the correct folding of most proteins requires the formation of disulfide bonds between cysteine residues to stabilize their structures. The formation of disulfide bonds is a reversible process that can be achieved through thiol–disulfide interchange. In eukaryotic cells, the folding of oxidative proteins is mainly catalyzed by a series of redox enzymes, such as protein disulfide isomerase (PDI), a member of the thioredoxin superfamily proteins [14]. Additionally, the folding of ER proteins is also dynamically regulated by the balance of the redox buffer, such as GSH/GSSG, which maintains the cellular redox state in a reduced state, GSH accounts for approximately 99% of the overall content, whereas it is found in only 50–60% in the endoplasmic reticulum [15]. PDI transfers two electrons to its substrate ERO1 (ER oxidoreductase 1), producing hydrogen peroxide. This electron transfer mode indicates the induction of ROS production [16].
Moreover, peroxisome, the organelles present in all eukaryotic cells, is believed to be the site to produce ROS. Peroxisome is closely related to the regulation of liver lipid homeostasis [17]. Medium- and long-chain fatty acids (FAs) are oxidized predominantly in mitochondria, whereas very long-chain FAs (VLCFAs) are metabolized almost exclusively by β-oxidation in the peroxisome and are involved in α-oxidation of fatty acids [18]. Peroxisomes produce acetyl coenzyme A through fatty acid β-oxidation, which plays a role in lipid signal transduction to promote lipid autophagy [19]. It is estimated that peroxisomes contribute about 35% of ROS in cells.
Moreover, NADPH oxidase, also known as NOX, is the only enzyme whose main function is to produce ROS. Researchers have identified seven isoforms, NOX1, NOX2, NOX3, NOX4, NOX5, DUOX1, and DUOX2, of which NOX2 is highly expressed in the liver [20]. NOX2 consists of 6 subunits: gp91phox, p22phox, p47phox, p67phox, p40phox, and Rac. Activation of NOX2 requires the translocation of p47phox, p67phox, p40phox, and Rac from the cytoplasm to the cell membrane, where they bind to gp91phox and p22phox to form the complex to produce the oxidants [21]. NOX2 affects signal transduction and immune function by transferring electrons from NADPH to molecular oxygen and generating large amounts of oxidants, a process involved in a variety of physiological activities such as cell proliferation and differentiation [22]. Studies have reported that in liver-resident Kupffer cells and infiltrating macrophages, NOX2-derived ROS stimulate them to produce pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β in response to a variety of factors, including oxidized low-density lipoproteins (LDL) and lipopolysaccharides (LPS) [23][24][25]. Therefore, over-activation of NOX2 leads to a surge of oxidants, which directly activates downstream inflammatory pathways and indirectly damages DNA and regulates protein phosphorylation, causing oxidative damage and exacerbating inflammation, which leads to cellular dysfunction and the onset and deterioration of diseases and affects the function and metabolism of tissues and organs [26][27]. Moreover, increased NOX2 activity has been shown in patients with NAFLD [28][29], suggesting a correlation between NOX2 and NAFLD. As mentioned above, oxidative stress serves as the “second hit” in the pathogenesis of NAFLD and is typically accompanied by a large amount of ROS. ROS can lead to the inactivation of mitochondrial respiratory chain enzymes and GAPDH, inhibit Ca2+ channels on the membrane, and induce liver cell damage [30]. Furthermore, ROS can induce the production of cytokines and Fas, along with their ligands, exacerbating lipid peroxidation, thus aggravating liver inflammation and hepatocyte fibrosis [31]. Cellular FFA loading can lead to changes in mitochondrial adaptability, enhancing fatty acid oxidation and upregulating electron transport, further resulting in excessive ROS production [32]. Thus, the excessive production of ROS can damage mitochondrial proteins, lipids, and DNA, a process that may play a role in the initiation of NASH. In the late stage of NASH, mitochondrial ultrastructure damage affects its function, reduces ATP synthesis, and intensifies ROS overproduction [33].

2.2. Thioredoxin (Trx) System

When the liver is exposed to high levels of ROS or electrophilic reagents, oxidative damage is prone to occur. At this point, the body initiates a series of antioxidant defense mechanisms [34]. The mammalian thiol-dependent antioxidant system comprises the thioredoxin (Trx) system and the GSH-glutaredoxin (Grx) system, both of which undergo interconversion between thiol (-SH) and disulfide bond (-S-S-) forms to eliminate ROS and maintain the redox homeostasis in the body [35].
The Trx system mainly consists of Trx, thioredoxin reductase (TrxR), and nicotinamide adenine dinucleotide phosphate (NADPH). It is a major thiol-dependent antioxidant system in the body, responsible for maintaining intracellular redox homeostasis and protecting cells from oxidative stress (Figure 1) [9][36]. Mammals have two isoforms of Trxs. Trx1 is present in the cytoplasm with five cysteines, two active site cysteines, and three structural cysteines, while Trx2 is located in the mitochondria with only two cysteine residues in its active center [36]. The Trx system transfers electrons to various key cellular proteins, including peroxiredoxin (Prx), which directly scavenges H2O2, and methionine-S-sulfoxide reductase (MsrA), which participates in oxidative stress
defense [35][37][38]. The proper functioning of the Trx system relies on TrxR accepting electrons from NADPH and transferring them to Trx. The activity and expression levels of TrxR directly determine the normal biological function of Trx and its downstream proteins. Mammalian cells have two forms of TrxR, cytoplasmic TrxR1 and mitochondrial TrxR2, both of which have FAD and NADPH binding domains as well as interface domains. At their N-terminus, they have an active site CVNVGC, and at their C-terminus, they possess a special sequence Gly-Cys-Sec-Gly (GCUG), with Sec being the catalytic active site of TrxR, which is necessary for its reduction activity [39].
Figure 1. The thioredoxin system plays a crucial role in modulating cell viability and proliferation. Thioredoxin can donate electrons to various enzymes, including peroxiredoxins, which have critical roles in cell signaling by either removing hydrogen peroxide or regulating redox-sensitive signaling molecules [40][41][42][43][44][45]. The redox state of thioredoxin can affect the function of several transcription factors, making it an important player in cellular signaling [46][47][48][49]. (ER: Endoplasmic reticulum, Keap1: kelch like ECH associated protein 1, Nrf2: Nuclear factor erythroid 2-related factor 2, ARE: Antioxidant response element, TrxR: Thioredoxin Reductase, Trx: Thioredoxin, Prx: Peroxiredoxin, NADPH: Nicotinamide Adenine Dinucleotide Phosphate).
Studies have shown significant changes in the Trx system in metabolic syndrome [50][51]. For example, in metabolic syndrome patients, consuming a diet rich in high-saturated fatty acids (HSFA) significantly increased the mRNA levels of Trx in adipose tissue. Simultaneously, postprandial adipose tissue TrxR1 mRNA significantly decreased.This suggests that the ability of Trx to revert from its oxidized form to its reduced form decreased, leading to a compensatory increase in Trx gene levels, indicating increased oxidative stress due to saturated fat intake [52]. The Trx system also plays a crucial role in adipocyte dysfunction and obesity [53]. In obese individuals with metabolic disorders, TrxR activity and Trx content in subcutaneous tissue are significantly increased because subcutaneous fat seems to provide good protection against increased oxidation in obese subjects compared to visceral fat. Protein levels of Trx, GPx, and CuZnSOD, as well as activities of GPx, GR, GST, TrxR, and SOD, were significantly higher in “at-risk” obese women than in metabolically healthy women [54]. While the expression of Trx-dependent peroxiredoxin (Prx3) in adipose tissue is significantly reduced [55], Prx3 is a key molecule regulating adipocyte oxidative stress and adipokine expression [56]. This may indicate that expression of various antioxidant enzymes is predominantly and specifically regulated by different transcription factors, but the detailed mechanism behind it needs further clarification.
Trx system in NASH has been shown to be changed in several studies [57][58]. Serum Trx levels are significantly increased in NASH patients compared to simple steatosis patients, making it a potential biomarker to distinguish NASH from early-stage fatty liver [59]. In animal models of choline-deficient liver steatosis, the activities of hepatic Trx1 and TrxR are upregulated at day 14 but significantly decreased at day 30 compared to day 14 [60]. Furthermore, the level of thioredoxin-interacting protein (TXNIP) in the liver of NAFLD patients is significantly elevated [61], and in mice fed a methionine and choline-deficient diet to induce NASH, the TXNIP gene was overexpressed, and expression of hepatic TrxR1 and TrxR2 decreased [62].

2.3. Glutathione (GSH)-Grx System

In addition to the Trx system, the glutathione-glutaredoxin (GSH-Grx) system is another crucial thiol-dependent redox system involved in cellular redox balance regulation [63]. It comprises NADPH, glutathione reductase (GR), and GSH coupled with Grxs, including cytosolic Grx1 and mitochondrial Grx2. The GSH-Grx systems mainly mediate redox signal transduction by reversibly catalyzing protein S-glutathionylation. GSH is the most abundant thiol small molecule, playing a vital role in maintaining intracellular redox homeostasis [64]. GSH can reduce various proteins, including glutathione peroxidases and glutathione transferases [65], and the resulting GSSG can be recycled by GR using NADPH as an electron donor [66].
Studies have shown that when Glrx1 KO mice are given a standard diet for some time, they develop obesity, hyperlipidemia, and fatty liver, resembling NAFLD. When fed a high-fat diet, Glrx1 KO mice develop fatty liver inflammation and progress to NASH [67]. In NAFLD animal models fed with a high-fat diet, S-glutathionylated protein levels increase, and an important target protein, Sirtuin-1 (SirT1), is identified. SirT1 is a NAD+-dependent class III histone deacetylase that regulates key transcription factors coordinating hepatic lipid metabolism [68][69][70]. Activation of SirT1 ameliorates Non-Alcoholic Fatty Liver (NAFL); conversely, hepatic SirT1 deficiency leads to steatosis [71], and thiol modification of SirT1 regulates lipid metabolism through acetylation of key transcription factors. S-glutathionylation inactivates SirT1 and promotes hyperacetylation and activation of downstream target proteins such as p53 and sterol regulatory element binding protein (SREBP) [68]. Activation of p53 is a cellular response to stress, resulting in the alteration of metabolism and the arrest of the cell cycle, and severe oxidative damage to hepatocytes may trigger p53 to induce cell death [72]. Studies also suggest that SirT1 overexpression can improve NAFLD [73], while Sirt1 knockout in mice leads to NAFLD (Figure 2) [74].
Figure 2. The GSH-Grx 1 system controls protein S-glutathionylation major players in the NAFLD. (GR: Glutathione Reductase, GSH: Glutathione, NADPH: Nicotinamide Adenine Dinucleotide Phosphate, Grx: Glutaredoxin, SirT-1: sirtuin type 1, PPAR: Peroxisome proliferators-activated receptors, PGC-1α: Peroxisome proliferator-activated receptor-gamma coactivator, SREBP-1c: Sterol regulatory element binding protein-1c).
Grx2 has two known isoforms encoded by the same gene (GLRX2): Grx2a (mitochondrial) and Grx2c (cytoplasmic) [75][76]. The role and effects of mitochondrial Grx2-mediated glutathionylation in cellular metabolism are still poorly understood. A mouse model with a depletion of mitochondrial Grx2 fed with a standard diet had spontaneous weight gain and accumulation of lipid droplets in the liver, suggesting that Grx2 is involved in mitochondrial redox environment and lipid metabolism regulation [77]. Genes such as cytochrome P-450 7a1 (Cyp7a1), which is specifically expressed in the liver and participates in the reduction of cholesterol, were found to be significantly downregulated in the mice model [77].

2.4. Nrf2 Signaling Pathway

The Keap1-Nrf2-ARE pathway is one of the major pathways that maintain cellular homeostasis during oxidative stress in the liver and is also regulated by Trx. Its function is to protect cells from endogenous and exogenous damage caused by oxidative stress. When Nrf2 accumulates significantly in the cell nucleus, a large number of antioxidant and phase II detoxification enzyme genes are upregulated [78]. Previous studies have identified over 200 downstream target genes regulated by Nrf2 in human cells [79], including the genes encoding NAD(P)H quinone oxidoreductase 1 (NQO1), HO-1, GST, GPx, CAT, SOD, and GR (Figure 1) [80]; It controls the expression of the glutamate–cysteine ligase catalytic subunit (GCLC) and glutamate–cysteine ligase modifier subunit (GCLM) to maintain the proper ratio of GSH to GSSG in cells [81]; Nrf2 also positively regulates some enzymes involved in liver detoxification, including Trx1, TrxR1, Gpx2, and GST [82][83]. These detoxification enzymes can eliminate H2O2, free radicals, and oxidized thiols in the cytoplasm, endoplasmic reticulum, and mitochondria [84]. Thus, Nrf2 represents a potential therapeutic target for NAFLD.
In addition to Nrf2, which has been shown to bind to the ARE of the Trx, TrxR, and Prx1 promoters [85][86][87], hypoxia-inducible factor (HIF)-1α synergistically up-regulates specific genes during hypoxia with another class of transcription factors, the E26 translationally specific (Ets) family of related proteins [88]. In human PC3 prostate cancer cells, the Prx1 gene promoter was significantly induced by either H2O2 or reoxygenation after hypoxic growth [89]. Co-transfection of constructs overexpressing Ets-1 or Ets-2 with the Prx1 promoter construct resulted in increased promoter activity, and ChIP assays also confirmed that both Ets-1 and Ets-2 bound to the Prx1 gene promoter in PC3 cells [89]. Thus, it can be hypothesized that the Ets pathway may play a role in regulating redox control during hypoxia and reoxygenation to complement the activation of the Nrf2 pathway. In addition, specificity protein 1 (Sp1), Fas/Jun, TATA box binding protein (TBP), cAMP response element binding protein (CREB), retinoic acid receptor/retinoid X receptor (RAR/RXR), and other transcription factors bind to their DNA sites to regulate Trx [85][87][90][91]. Retinoid X receptor (RAR/RXR) and other transcription factors that bind to DNA sites to regulate Trx; Octamer binding protein (Oct-1), Sp1, Sp3, and other transcription factors bind to the DNA site to regulate TrxR [92]. However, whether these transcriptional regulations are involved in NAFLD/NASH is unclear.

References

  1. Stefan, N.; Häring, H.U.; Cusi, K. Non-alcoholic fatty liver disease: Causes, diagnosis, cardiometabolic consequences, and treatment strategies. Lancet. Diabetes Endocrinol. 2019, 7, 313–324.
  2. Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922.
  3. Eslam, M.; Sanyal, A.J.; George, J. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014.
  4. Seghieri, M.; Christensen, A.S.; Andersen, A.; Solini, A.; Knop, F.K.; Vilsbøll, T. Future Perspectives on GLP-1 Receptor Agonists and GLP-1/glucagon Receptor Co-agonists in the Treatment of NAFLD. Front. Endocrinol. 2018, 9, 649.
  5. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metab. Clin. Exp. 2016, 65, 1038–1048.
  6. Tilg, H.; Moschen, A.R. Evolution of inflammation in nonalcoholic fatty liver disease: The multiple parallel hits hypothesis. Hepatology 2010, 52, 1836–1846.
  7. Zarkovic, N. Roles and Functions of ROS and RNS in Cellular Physiology and Pathology. Cells 2020, 9, 767.
  8. Lushchak, V.I. Free radicals, reactive oxygen species, oxidative stress and its classification. Chem. Biol. Interact. 2014, 224, 164–175.
  9. Nordberg, J.; Arnér, E.S. Reactive oxygen species, antioxidants, and the mammalian thioredoxin system. Free. Radic. Biol. Med. 2001, 31, 1287–1312.
  10. Imlay, J.A.; Chin, S.M.; Linn, S. Toxic DNA damage by hydrogen peroxide through the Fenton reaction in vivo and in vitro. Science 1988, 240, 640–642.
  11. Shabalina, I.G.; Nedergaard, J. Mitochondrial (‘mild’) uncoupling and ROS production: Physiologically relevant or not? Biochem. Soc. Trans. 2011, 39, 1305–1309.
  12. Liu, X.; Zhang, J.; Ming, Y.; Chen, X.; Zeng, M.; Mao, Y. The aggravation of mitochondrial dysfunction in nonalcoholic fatty liver disease accompanied with type 2 diabetes mellitus. Scand. J. Gastroenterol. 2015, 50, 1152–1159.
  13. García-Berumen, C.I.; Ortiz-Avila, O.; Vargas-Vargas, M.A.; Del Rosario-Tamayo, B.A.; Guajardo-López, C.; Saavedra-Molina, A.; Rodríguez-Orozco, A.R.; Cortés-Rojo, C. The severity of rat liver injury by fructose and high fat depends on the degree of respiratory dysfunction and oxidative stress induced in mitochondria. Lipids Health Dis. 2019, 18, 78.
  14. Okumura, M.; Noi, K.; Kanemura, S.; Kinoshita, M.; Saio, T.; Inoue, Y.; Hikima, T.; Akiyama, S.; Ogura, T.; Inaba, K. Dynamic assembly of protein disulfide isomerase in catalysis of oxidative folding. Nat. Chem. Biol. 2019, 15, 499–509.
  15. Rashdan, N.A.; Pattillo, C.B. Hydrogen peroxide in the ER: A tale of triage. Redox Biol. 2020, 28, 101358.
  16. Zeeshan, H.M.; Lee, G.H.; Kim, H.R.; Chae, H.J. Endoplasmic Reticulum Stress and Associated ROS. Int. J. Mol. Sci. 2016, 17, 327.
  17. Ding, L.; Sun, W.; Balaz, M.; He, A.; Klug, M.; Wieland, S.; Caiazzo, R.; Raverdy, V.; Pattou, F.; Lefebvre, P.; et al. Peroxisomal β-oxidation acts as a sensor for intracellular fatty acids and regulates lipolysis. Nat. Metab. 2021, 3, 1648–1661.
  18. Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive Oxygen Species in Metabolic and Inflammatory Signaling. Circ. Res. 2018, 122, 877–902.
  19. Zhang, J.; Tripathi, D.N.; Jing, J.; Alexander, A.; Kim, J.; Powell, R.T.; Dere, R.; Tait-Mulder, J.; Lee, J.H.; Paull, T.T.; et al. ATM functions at the peroxisome to induce pexophagy in response to ROS. Nat. Cell Biol. 2015, 17, 1259–1269.
  20. Zhang, Y.; Murugesan, P.; Huang, K.; Cai, H. NADPH oxidases and oxidase crosstalk in cardiovascular diseases: Novel therapeutic targets. Nat. Rev. Cardiol. 2020, 17, 170–194.
  21. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313.
  22. Kim, S.Y.; Jeong, J.M.; Kim, S.J.; Seo, W.; Kim, M.H.; Choi, W.M.; Yoo, W.; Lee, J.H.; Shim, Y.R.; Yi, H.S.; et al. Pro-inflammatory hepatic macrophages generate ROS through NADPH oxidase 2 via endocytosis of monomeric TLR4-MD2 complex. Nat. Commun. 2017, 8, 2247.
  23. Thakur, V.; Pritchard, M.T.; McMullen, M.R.; Wang, Q.; Nagy, L.E. Chronic ethanol feeding increases activation of NADPH oxidase by lipopolysaccharide in rat Kupffer cells: Role of increased reactive oxygen in LPS-stimulated ERK1/2 activation and TNF-alpha production. J. Leukoc. Biol. 2006, 79, 1348–1356.
  24. Meng, T.; Yu, J.; Lei, Z.; Wu, J.; Wang, S.; Bo, Q.; Zhang, X.; Ma, Z.; Yu, J. Propofol reduces lipopolysaccharide-induced, NADPH oxidase (NOX 2) mediated TNF-α and IL-6 production in macrophages. Clin. Dev. Immunol. 2013, 2013, 325481.
  25. Bae, Y.S.; Lee, J.H.; Choi, S.H.; Kim, S.; Almazan, F.; Witztum, J.L.; Miller, Y.I. Macrophages generate reactive oxygen species in response to minimally oxidized low-density lipoprotein: Toll-like receptor 4- and spleen tyrosine kinase-dependent activation of NADPH oxidase 2. Circ. Res. 2009, 104, 210–218.
  26. Yang, C.-S.; Kim, J.-J.; Lee, S.J.; Hwang, J.H.; Lee, C.-H.; Lee, M.-S.; Jo, E.-K. TLR3-Triggered Reactive Oxygen Species Contribute to Inflammatory Responses by Activating Signal Transducer and Activator of Transcription-1. J. Immunol. 2013, 190, 6368–6377.
  27. Musso, G.; Cassader, M.; Paschetta, E.; Gambino, R. Bioactive Lipid Species and Metabolic Pathways in Progression and Resolution of Nonalcoholic Steatohepatitis. Gastroenterology 2018, 155, 282–302.e8.
  28. Loffredo, L.; Zicari, A.M.; Perri, L.; Carnevale, R.; Nocella, C.; Angelico, F.; Del Ben, M.; Mosca, A.; Zaffina, S.; Panera, N.; et al. Does Nox2 Overactivate in Children with Nonalcoholic Fatty Liver Disease? Antioxid. Redox Signal. 2019, 30, 1325–1330.
  29. Grossini, E.; Garhwal, D.P.; Calamita, G.; Romito, R.; Rigamonti, C.; Minisini, R.; Smirne, C.; Surico, D.; Bellan, M.; Pirisi, M. Exposure to Plasma From Non-alcoholic Fatty Liver Disease Patients Affects Hepatocyte Viability, Generates Mitochondrial Dysfunction, and Modulates Pathways Involved in Fat Accumulation and Inflammation. Front. Med. 2021, 8, 693997.
  30. Wang, J.; He, W.; Tsai, P.J.; Chen, P.H.; Ye, M.; Guo, J.; Su, Z. Mutual interaction between endoplasmic reticulum and mitochondria in nonalcoholic fatty liver disease. Lipids Health Dis. 2020, 19, 72.
  31. Pessayre, D.; Berson, A.; Fromenty, B.; Mansouri, A. Mitochondria in steatohepatitis. Semin. Liver Dis. 2001, 21, 57–69.
  32. Du, X.; Edelstein, D.; Obici, S.; Higham, N.; Zou, M.H.; Brownlee, M. Insulin resistance reduces arterial prostacyclin synthase and eNOS activities by increasing endothelial fatty acid oxidation. J. Clin. Investig. 2006, 116, 1071–1080.
  33. Arab, J.P.; Arrese, M.; Trauner, M. Recent Insights into the Pathogenesis of Nonalcoholic Fatty Liver Disease. Annu. Rev. Pathol. 2018, 13, 321–350.
  34. Abe, Y.; Hines, I.N.; Zibari, G.; Pavlick, K.; Gray, L.; Kitagawa, Y.; Grisham, M.B. Mouse model of liver ischemia and reperfusion injury: Method for studying reactive oxygen and nitrogen metabolites in vivo. Free. Radic. Biol. Med. 2009, 46, 1–7.
  35. Holmgren, A. Thioredoxin. Annu. Rev. Biochem. 1985, 54, 237–271.
  36. Arnér, E.S.; Holmgren, A. Physiological functions of thioredoxin and thioredoxin reductase. Eur. J. Biochem. 2000, 267, 6102–6109.
  37. Lu, J.; Holmgren, A. Thioredoxin system in cell death progression. Antioxid. Redox Signal. 2012, 17, 1738–1747.
  38. Zhong, L.; Arnér, E.S.; Holmgren, A. Structure and mechanism of mammalian thioredoxin reductase: The active site is a redox-active selenolthiol/selenenylsulfide formed from the conserved cysteine-selenocysteine sequence. Proc. Natl. Acad. Sci. USA 2000, 97, 5854–5859.
  39. Lu, J.; Holmgren, A. Selenoproteins. J. Biol. Chem. 2009, 284, 723–727.
  40. Lillig, C.H.; Holmgren, A. Thioredoxin and related molecules–from biology to health and disease. Antioxid. Redox Signal. 2007, 9, 25–47.
  41. Gladyshev, V.N.; Jeang, K.-T.; Stadtman, T.C. Selenocysteine, identified as the penultimate C-terminal residue in human T-cell thioredoxin reductase, corresponds to TGA in the human placental gene. Proc. Natl. Acad. Sci. USA 1996, 93, 6146–6151.
  42. Zhong, L.; Arnér, E.S.; Ljung, J.; Åslund, F.; Holmgren, A. Rat and calf thioredoxin reductase are homologous to glutathione reductase with a carboxyl-terminal elongation containing a conserved catalytically active penultimate selenocysteine residue. J. Biol. Chem. 1998, 273, 8581–8591.
  43. Lee, S.-R.; Kim, J.-R.; Kwon, K.-S.; Yoon, H.W.; Levine, R.L.; Ginsburg, A.; Rhee, S.G. Molecular cloning and characterization of a mitochondrial selenocysteine-containing thioredoxin reductase from rat liver. J. Biol. Chem. 1999, 274, 4722–4734.
  44. Rigobello, M.P.; Callegaro, M.T.; Barzon, E.; Benetti, M.; Bindoli, A. Purification of mitochondrial thioredoxin reductase and its involvement in the redox regulation of membrane permeability. Free Radic. Biol. Med. 1998, 24, 370–376.
  45. Rhee, S.G.; Chae, H.Z.; Kim, K. Peroxiredoxins: A historical overview and speculative preview of novel mechanisms and emerging concepts in cell signaling. Free Radic. Biol. Med. 2005, 38, 1543–1552.
  46. Morel, Y.; Barouki, R. Repression of gene expression by oxidative stress. Biochem. J. 1999, 342, 481–496.
  47. Welsh, S.J.; Bellamy, W.T.; Briehl, M.M.; Powis, G. The redox protein thioredoxin-1 (Trx-1) increases hypoxia-inducible factor 1α protein expression: Trx-1 overexpression results in increased vascular endothelial growth factor production and enhanced tumor angiogenesis. Cancer Res. 2002, 62, 5089–5095.
  48. Makino, Y.; Yoshikawa, N.; Okamoto, K.; Hirota, K.; Yodoi, J.; Makino, I.; Tanaka, H. Direct association with thioredoxin allows redox regulation of glucocorticoid receptor function. J. Biol. Chem. 1999, 274, 3182–3188.
  49. Ueno, M.; Masutani, H.; Arai, R.J.; Yamauchi, A.; Hirota, K.; Sakai, T.; Inamoto, T.; Yamaoka, Y.; Yodoi, J.; Nikaido, T. Thioredoxin-dependent redox regulation of p53-mediated p21 activation. J. Biol. Chem. 1999, 274, 35809–35815.
  50. Crunkhorn, S. Cardiovascular disease: Thioredoxin lowers hypertension. Nat. Rev. Drug Discov. 2017, 16, 240.
  51. Kaimul, A.M.; Nakamura, H.; Masutani, H.; Yodoi, J. Thioredoxin and thioredoxin-binding protein-2 in cancer and metabolic syndrome. Free Radic. Biol. Med. 2007, 43, 861–868.
  52. Peña-Orihuela, P.; Camargo, A.; Rangel-Zuñiga, O.A.; Perez-Martinez, P.; Cruz-Teno, C.; Delgado-Lista, J.; Yubero-Serrano, E.M.; Paniagua, J.A.; Tinahones, F.J.; Malagon, M.M.; et al. Antioxidant system response is modified by dietary fat in adipose tissue of metabolic syndrome patients. J. Nutr. Biochem. 2013, 24, 1717–1723.
  53. Jankovic, A.; Korac, A.; Buzadzic, B.; Otasevic, V.; Stancic, A.; Daiber, A.; Korac, B. Redox implications in adipose tissue (dys)function--A new look at old acquaintances. Redox Biol. 2015, 6, 19–32.
  54. Jankovic, A.; Korac, A.; Srdic-Galic, B.; Buzadzic, B.; Otasevic, V.; Stancic, A.; Vucetic, M.; Markelic, M.; Velickovic, K.; Golic, I.; et al. Differences in the redox status of human visceral and subcutaneous adipose tissues--relationships to obesity and metabolic risk. Metab. Clin. Exp. 2014, 63, 661–671.
  55. Bouwman, F.G.; Claessens, M.; van Baak, M.A.; Noben, J.P.; Wang, P.; Saris, W.H.; Mariman, E.C. The physiologic effects of caloric restriction are reflected in the in vivo adipocyte-enriched proteome of overweight/obese subjects. J. Proteome Res. 2009, 8, 5532–5540.
  56. Huh, J.Y.; Kim, Y.; Jeong, J.; Park, J.; Kim, I.; Huh, K.H.; Kim, Y.S.; Woo, H.A.; Rhee, S.G.; Lee, K.J.; et al. Peroxiredoxin 3 is a key molecule regulating adipocyte oxidative stress, mitochondrial biogenesis, and adipokine expression. Antioxid. Redox Signal. 2012, 16, 229–243.
  57. Okuyama, H.; Son, A.; Ahsan, M.K.; Masutani, H.; Nakamura, H.; Yodoi, J. Thioredoxin and thioredoxin binding protein 2 in the liver. IUBMB life 2008, 60, 656–660.
  58. Kim, C.H.; Younossi, Z.M. Nonalcoholic fatty liver disease: A manifestation of the metabolic syndrome. Clevel. Clin. J. Med. 2008, 75, 721–728.
  59. Sumida, Y.; Nakashima, T.; Yoh, T.; Furutani, M.; Hirohama, A.; Kakisaka, Y.; Nakajima, Y.; Ishikawa, H.; Mitsuyoshi, H.; Okanoue, T.; et al. Serum thioredoxin levels as a predictor of steatohepatitis in patients with nonalcoholic fatty liver disease. J. Hepatol. 2003, 38, 32–38.
  60. Grattagliano, I.; Caraceni, P.; Calamita, G.; Ferri, D.; Gargano, I.; Palasciano, G.; Portincasa, P. Severe liver steatosis correlates with nitrosative and oxidative stress in rats. Eur. J. Clin. Investig. 2008, 38, 523–530.
  61. Park, B.J.; Cha, M.K.; Kim, I.H. Thioredoxin 1 as a serum marker for ovarian cancer and its use in combination with CA125 for improving the sensitivity of ovarian cancer diagnoses. Biomark. Biochem. Indic. Expo. Response Susceptibility Chem. 2014, 19, 604–610.
  62. Gornicka, A.; Morris-Stiff, G.; Thapaliya, S.; Papouchado, B.G.; Berk, M.; Feldstein, A.E. Transcriptional profile of genes involved in oxidative stress and antioxidant defense in a dietary murine model of steatohepatitis. Antioxid. Redox Signal. 2011, 15, 437–445.
  63. Holmgren, A. Thioredoxin and Glutaredoxin Systems. J. Biol. Chem. 1989, 264, 13963–13966.
  64. Morgan, B.; Ezerina, D.; Amoako, T.N.E.; Riemer, J.; Seedorf, M.; Dick, T.P. Multiple glutathione disulfide removal pathways mediate cytosolic redox homeostasis. Nat. Chem. Biol. 2013, 9, 119–125.
  65. Diaz-Vivancos, P.; de Simone, A.; Kiddie, G.; Foyer, C.H. Glutathione—Linking cell proliferation to oxidative stress. Free Radic. Biol. Med. 2015, 89, 1154–1164.
  66. Lillig, C.H.; Berndt, C.; Holmgren, A. Glutaredoxin systems. Bba-Gen. Subj. 2008, 1780, 1304–1317.
  67. Shao, D.; Han, J.; Hou, X.; Fry, J.; Behring, J.B.; Seta, F.; Long, M.T.; Roy, H.K.; Cohen, R.A.; Matsui, R.; et al. Glutaredoxin-1 Deficiency Causes Fatty Liver and Dyslipidemia by Inhibiting Sirtuin-1. Antioxid. Redox Signal. 2017, 27, 313–327.
  68. Ponugoti, B.; Kim, D.H.; Xiao, Z.; Smith, Z.; Miao, J.; Zang, M.; Wu, S.Y.; Chiang, C.M.; Veenstra, T.D.; Kemper, J.K. SIRT1 deacetylates and inhibits SREBP-1C activity in regulation of hepatic lipid metabolism. J. Biol. Chem. 2010, 285, 33959–33970.
  69. Picard, F.; Kurtev, M.; Chung, N.; Topark-Ngarm, A.; Senawong, T.; Machado De Oliveira, R.; Leid, M.; McBurney, M.W.; Guarente, L. Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature 2004, 429, 771–776.
  70. Haigis, M.C.; Sinclair, D.A. Mammalian sirtuins: Biological insights and disease relevance. Annu. Rev. Pathol. 2010, 5, 253–295.
  71. Pfluger, P.T.; Herranz, D.; Velasco-Miguel, S.; Serrano, M.; Tschöp, M.H. Sirt1 protects against high-fat diet-induced metabolic damage. Proc. Natl. Acad. Sci. USA 2008, 105, 9793–9798.
  72. Shao, D.; Fry, J.L.; Han, J.; Hou, X.; Pimentel, D.R.; Matsui, R.; Cohen, R.A.; Bachschmid, M.M. A redox-resistant sirtuin-1 mutant protects against hepatic metabolic and oxidant stress. J. Biol. Chem. 2014, 289, 7293–7306.
  73. Deng, X.Q.; Chen, L.L.; Li, N.X. The expression of SIRT1 in nonalcoholic fatty liver disease induced by high-fat diet in rats. Liver Int. Off. J. Int. Assoc. Study Liver 2007, 27, 708–715.
  74. Yin, H.; Hu, M.; Liang, X.; Ajmo, J.M.; Li, X.; Bataller, R.; Odena, G.; Stevens, S.M., Jr.; You, M. Deletion of SIRT1 from hepatocytes in mice disrupts lipin-1 signaling and aggravates alcoholic fatty liver. Gastroenterology 2014, 146, 801–811.
  75. Hudemann, C.; Lönn, M.E.; Godoy, J.R.; Zahedi Avval, F.; Capani, F.; Holmgren, A.; Lillig, C.H. Identification, expression pattern, and characterization of mouse glutaredoxin 2 isoforms. Antioxid. Redox Signal. 2009, 11, 1–14.
  76. Lönn, M.E.; Hudemann, C.; Berndt, C.; Cherkasov, V.; Capani, F.; Holmgren, A.; Lillig, C.H. Expression pattern of human glutaredoxin 2 isoforms: Identification and characterization of two testis/cancer cell-specific isoforms. Antioxid. Redox Signal. 2008, 10, 547–557.
  77. Scalcon, V.; Folda, A.; Lupo, M.G.; Tonolo, F.; Pei, N.; Battisti, I.; Ferri, N.; Arrigoni, G.; Bindoli, A.; Holmgren, A.; et al. Mitochondrial depletion of glutaredoxin 2 induces metabolic dysfunction-associated fatty liver disease in mice. Redox Biol. 2022, 51, 102277.
  78. Yamamoto, M.; Kensler, T.W.; Motohashi, H. The KEAP1-NRF2 System: A Thiol-Based Sensor-Effector Apparatus for Maintaining Redox Homeostasis. Physiol. Rev. 2018, 98, 1169–1203.
  79. Malhotra, D.; Portales-Casamar, E.; Singh, A.; Srivastava, S.; Arenillas, D.; Happel, C.; Shyr, C.; Wakabayashi, N.; Kensler, T.W.; Wasserman, W.W.; et al. Global mapping of binding sites for Nrf2 identifies novel targets in cell survival response through ChIP-Seq profiling and network analysis. Nucleic Acids Res. 2010, 38, 5718–5734.
  80. Galicia-Moreno, M.; Lucano-Landeros, S.; Monroy-Ramirez, H.C.; Silva-Gomez, J.; Gutierrez-Cuevas, J.; Santos, A.; Armendariz-Borunda, J. Roles of Nrf2 in Liver Diseases: Molecular, Pharmacological, and Epigenetic Aspects. Antioxidants 2020, 9, 980.
  81. Higgins, L.G.; Kelleher, M.O.; Eggleston, I.M.; Itoh, K.; Yamamoto, M.; Hayes, J.D. Transcription factor Nrf2 mediates an adaptive response to sulforaphane that protects fibroblasts in vitro against the cytotoxic effects of electrophiles, peroxides and redox-cycling agents. Toxicol. Appl. Pharmacol. 2009, 237, 267–280.
  82. Hawkes, H.J.; Karlenius, T.C.; Tonissen, K.F. Regulation of the human thioredoxin gene promoter and its key substrates: A study of functional and putative regulatory elements. Biochim. Biophys. Acta 2014, 1840, 303–314.
  83. Harvey, C.J.; Thimmulappa, R.K.; Singh, A.; Blake, D.J.; Ling, G.; Wakabayashi, N.; Fujii, J.; Myers, A.; Biswal, S. Nrf2-regulated glutathione recycling independent of biosynthesis is critical for cell survival during oxidative stress. Free Radic. Biol. Med. 2009, 46, 443–453.
  84. Hayes, J.D.; Dinkova-Kostova, A.T. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem. Sci. 2014, 39, 199–218.
  85. Kim, Y.C.; Masutani, H.; Yamaguchi, Y.; Itoh, K.; Yamamoto, M.; Yodoi, J. Hemin-induced activation of the thioredoxin gene by Nrf2. A differential regulation of the antioxidant responsive element by a switch of its binding factors. J. Biol. Chem. 2001, 276, 18399–18406.
  86. Chorley, B.N.; Campbell, M.R.; Wang, X.; Karaca, M.; Sambandan, D.; Bangura, F.; Xue, P.; Pi, J.; Kleeberger, S.R.; Bell, D.A. Identification of novel NRF2-regulated genes by ChIP-Seq: Influence on retinoid X receptor alpha. Nucleic Acids Res. 2012, 40, 7416–7429.
  87. Yodoi, J.; Nakamura, H.; Masutani, H. Redox regulation of stress signals: Possible roles of dendritic stellate TRX producer cells (DST cell types). Biol. Chem. 2002, 383, 585–590.
  88. Charlot, C.; Dubois-Pot, H.; Serchov, T.; Tourrette, Y.; Wasylyk, B. A review of post-translational modifications and subcellular localization of Ets transcription factors: Possible connection with cancer and involvement in the hypoxic response. Methods Mol. Biol. 2010, 647, 3–30.
  89. Shiota, M.; Izumi, H.; Miyamoto, N.; Onitsuka, T.; Kashiwagi, E.; Kidani, A.; Hirano, G.; Takahashi, M.; Ono, M.; Kuwano, M.; et al. Ets regulates peroxiredoxin1 and 5 expressions through their interaction with the high-mobility group protein B1. Cancer Sci. 2008, 99, 1950–1959.
  90. Osborne, S.A.; Hawkes, H.J.; Baldwin, B.L.; Alexander, K.A.; Svingen, T.; Clarke, F.M.; Tonissen, K.F. The tert-butylhydroquinone-mediated activation of the human thioredoxin gene reveals a novel promoter structure. Biochem. J. 2006, 398, 269–277.
  91. Bai, J.; Nakamura, H.; Kwon, Y.W.; Hattori, I.; Yamaguchi, Y.; Kim, Y.C.; Kondo, N.; Oka, S.; Ueda, S.; Masutani, H.; et al. Critical roles of thioredoxin in nerve growth factor-mediated signal transduction and neurite outgrowth in PC12 cells. J. Neurosci. Off. J. Soc. Neurosci. 2003, 23, 503–509.
  92. Rundlöf, A.K.; Carlsten, M.; Arnér, E.S. The core promoter of human thioredoxin reductase 1: Cloning, transcriptional activity, and Oct-1, Sp1, and Sp3 binding reveal a housekeeping-type promoter for the AU-rich element-regulated gene. J. Biol. Chem. 2001, 276, 30542–30551.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 133
Revisions: 2 times (View History)
Update Date: 11 Sep 2023
1000/1000
ScholarVision Creations