Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 611
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 85: 485-514, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27145839

ABSTRACT

Radical S-adenosylmethionine (SAM) enzymes catalyze an astonishing array of complex and chemically challenging reactions across all domains of life. Of approximately 114,000 of these enzymes, 8 are known to be present in humans: MOCS1, molybdenum cofactor biosynthesis; LIAS, lipoic acid biosynthesis; CDK5RAP1, 2-methylthio-N(6)-isopentenyladenosine biosynthesis; CDKAL1, methylthio-N(6)-threonylcarbamoyladenosine biosynthesis; TYW1, wybutosine biosynthesis; ELP3, 5-methoxycarbonylmethyl uridine; and RSAD1 and viperin, both of unknown function. Aberrations in the genes encoding these proteins result in a variety of diseases. In this review, we summarize the biochemical characterization of these 8 radical S-adenosylmethionine enzymes and, in the context of human health, describe the deleterious effects that result from such genetic mutations.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Heart Defects, Congenital/genetics , Metal Metabolism, Inborn Errors/genetics , Mutation , Neurodegenerative Diseases/genetics , S-Adenosylmethionine/metabolism , Carbon-Carbon Lyases , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/pathology , Gene Expression , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/pathology , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Metal Metabolism, Inborn Errors/enzymology , Metal Metabolism, Inborn Errors/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxidoreductases Acting on CH-CH Group Donors , Proteins/genetics , Proteins/metabolism , Thioctic Acid/metabolism , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism
2.
Cell ; 159(7): 1615-25, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25525879

ABSTRACT

Sirtuins (SIRTs) are critical enzymes that govern genome regulation, metabolism, and aging. Despite conserved deacetylase domains, mitochondrial SIRT4 and SIRT5 have little to no deacetylase activity, and a robust catalytic activity for SIRT4 has been elusive. Here, we establish SIRT4 as a cellular lipoamidase that regulates the pyruvate dehydrogenase complex (PDH). Importantly, SIRT4 catalytic efficiency for lipoyl- and biotinyl-lysine modifications is superior to its deacetylation activity. PDH, which converts pyruvate to acetyl-CoA, has been known to be primarily regulated by phosphorylation of its E1 component. We determine that SIRT4 enzymatically hydrolyzes the lipoamide cofactors from the E2 component dihydrolipoyllysine acetyltransferase (DLAT), diminishing PDH activity. We demonstrate SIRT4-mediated regulation of DLAT lipoyl levels and PDH activity in cells and in vivo, in mouse liver. Furthermore, metabolic flux switching via glutamine stimulation induces SIRT4 lipoamidase activity to inhibit PDH, highlighting SIRT4 as a guardian of cellular metabolism.


Subject(s)
Mitochondrial Proteins/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Sirtuins/metabolism , Amidohydrolases/metabolism , Animals , Gene Knockdown Techniques , Glutamine/metabolism , Humans , Liver/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Rats , Sirtuins/genetics , Thioctic Acid/analogs & derivatives , Thioctic Acid/metabolism
3.
PLoS Biol ; 21(6): e3002177, 2023 06.
Article in English | MEDLINE | ID: mdl-37368881

ABSTRACT

Lipoic acid is an essential biomolecule found in all domains of life and is involved in central carbon metabolism and dissimilatory sulfur oxidation. The machineries for lipoate assembly in mitochondria and chloroplasts of higher eukaryotes, as well as in the apicoplasts of some protozoa, are all of prokaryotic origin. Here, we provide experimental evidence for a novel lipoate assembly pathway in bacteria based on a sLpl(AB) lipoate:protein ligase, which attaches octanoate or lipoate to apo-proteins, and 2 radical SAM proteins, LipS1 and LipS2, which work together as lipoyl synthase and insert 2 sulfur atoms. Extensive homology searches combined with genomic context analyses allowed us to precisely distinguish between the new and established pathways and map them on the tree of life. This not only revealed a much wider distribution of lipoate biogenesis systems than expected, in particular, the novel sLpl(AB)-LipS1/S2 pathway, and indicated a highly modular nature of the enzymes involved, with unforeseen combinations, but also provided a new framework for the evolution of lipoate assembly. Our results show that dedicated machineries for both de novo lipoate biogenesis and scavenging from the environment were implemented early in evolution and that their distribution in the 2 prokaryotic domains was shaped by a complex network of horizontal gene transfers, acquisition of additional genes, fusions, and losses. Our large-scale phylogenetic analyses identify the bipartite archaeal LplAB ligase as the ancestor of the bacterial sLpl(AB) proteins, which were obtained by horizontal gene transfer. LipS1/S2 have a more complex evolutionary history with multiple of such events but probably also originated in the domain archaea.


Subject(s)
Thioctic Acid , Thioctic Acid/genetics , Thioctic Acid/metabolism , Peptide Synthases/genetics , Peptide Synthases/metabolism , Phylogeny , Bacterial Proteins/metabolism , Sulfur
4.
Am J Pathol ; 194(2): 307-320, 2024 02.
Article in English | MEDLINE | ID: mdl-38245252

ABSTRACT

Sleep deprivation (SD) is a global public health burden, and has a detrimental role in the nervous system. Retina is an important part of the central nervous system; however, whether SD affects retinal structures and functions remains largely unknown. Herein, chronic SD mouse model indicated that loss of sleep for 4 months could result in reductions in the visual functions, but without obvious morphologic changes of the retina. Ultrastructural analysis by transmission electron microscope revealed the deterioration of mitochondria, which was accompanied with the decrease of multiple mitochondrial proteins in the retina. Mechanistically, oxidative stress was provoked by chronic SD, which could be ameliorated after rest, and thus restore retinal homeostasis. Moreover, the supplementation of two antioxidants, α-lipoic acid and N-acetyl-l-cysteine, could reduce retinal reactive oxygen species, repair damaged mitochondria, and, as a result, improve the retinal functions. Overall, this work demonstrated the essential roles of sleep in maintaining the integrity and health of the retina. More importantly, it points towards supplementation of antioxidants as an effective intervention strategy for people experiencing sleep shortages.


Subject(s)
Sleep Deprivation , Thioctic Acid , Humans , Mice , Animals , Sleep Deprivation/complications , Sleep Deprivation/metabolism , Oxidative Stress/physiology , Antioxidants/pharmacology , Retina/metabolism , Thioctic Acid/pharmacology , Thioctic Acid/metabolism
5.
J Biol Chem ; 299(10): 105228, 2023 10.
Article in English | MEDLINE | ID: mdl-37703993

ABSTRACT

The enzyme cofactor (R)-lipoic acid plays a critical role in central carbon metabolism due to its catalytic function in the generation of acetyl-CoA, which links glycolysis with the tricarboxylic acid cycle. This cofactor is also essential for the generation of succinyl CoA within the tricarboxylic acid cycle. However, the biological functions of (R)-lipoic acid extend beyond metabolism owing to its facile redox chemistry. Most recently, the reduced form of (R)-lipoic acid, (R)-dihydrolipoic acid, has been shown to inhibit histone deacetylases (HDACs) with selectivity for the inhibition of HDAC6. Here, we report the 2.4 Å-resolution X-ray crystal structure of the complex between (R)-dihydrolipoic acid and HDAC6 catalytic domain 2 from Danio rerio, and we report a dissociation constant (KD) of 350 nM for this complex as determined by isothermal titration calorimetry. The crystal structure illuminates key affinity determinants in the enzyme active site, including thiolate-Zn2+ coordination and S-π interactions in the F583-F643 aromatic crevice. This study provides the first visualization of the connection between HDAC function and the biological response to oxidative stress: the dithiol moiety of (R)-dihydrolipoic acid can serve as a redox-regulated pharmacophore capable of simultaneously targeting the catalytic Zn2+ ion and the aromatic crevice in the active site of HDAC6.


Subject(s)
Thioctic Acid , Animals , Histone Deacetylase 6/metabolism , Thioctic Acid/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Zebrafish/metabolism
6.
J Biol Chem ; 299(9): 105075, 2023 09.
Article in English | MEDLINE | ID: mdl-37481209

ABSTRACT

Iron-sulfur clusters (ISC) are essential cofactors that participate in electron transfer, environmental sensing, and catalysis. Amongst the most ancient ISC-containing proteins are the ferredoxin (FDX) family of electron carriers. Humans have two FDXs- FDX1 and FDX2, both of which are localized to mitochondria, and the latter of which is itself important for ISC synthesis. We have previously shown that hypoxia can eliminate the requirement for some components of the ISC biosynthetic pathway, but FDXs were not included in that study. Here, we report that FDX1, but not FDX2, is dispensable under 1% O2 in cultured human cells. We find that FDX1 is essential for production of the lipoic acid cofactor, which is synthesized by the ISC-containing enzyme lipoyl synthase. While hypoxia can rescue the growth phenotype of either FDX1 or lipoyl synthase KO cells, lipoylation in these same cells is not rescued, arguing against an alternative biosynthetic route or salvage pathway for lipoate in hypoxia. Our work reveals the divergent roles of FDX1 and FDX2 in mitochondria, identifies a role for FDX1 in lipoate synthesis, and suggests that loss of lipoic acid can be tolerated under low oxygen tensions in cell culture.


Subject(s)
Ferredoxins , Lipoylation , Humans , Ferredoxins/genetics , Ferredoxins/metabolism , Thioctic Acid/metabolism , Cell Hypoxia/drug effects , Gene Knockout Techniques , Oxygen/pharmacology , Proteome/drug effects , Proteome/genetics , Sulfurtransferases/genetics , Sulfurtransferases/metabolism , Binding Sites , Protein Stability , Protein Biosynthesis/drug effects
7.
J Transl Med ; 22(1): 82, 2024 01 20.
Article in English | MEDLINE | ID: mdl-38245790

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a liver disorder characterized by the ac-cumulation of fat in hepatocytes without alcohol consumption. Mitochondrial dysfunction and endoplasmic reticulum (ER) stress play significant roles in NAFLD pathogenesis. The unfolded protein response in mitochondria (UPRmt) is an adaptive mechanism that aims to restore mitochondrial protein homeostasis and mitigate cellular stress. This study aimed to investigate the effects of ( +)-Lipoic acid (ALA) on UPRmt, inflammation, and oxidative stress in an in vitro model of NAFLD using HepG2 cells treated with palmitic acid and oleic acid to induce steatosis. RESULTS: Treatment with palmitic and oleic acids increased UPRmt-related proteins HSP90 and HSP60 (heat shock protein), and decreased CLPP (caseinolytic protease P), indicating ER stress activation. ALA treatment at 1 µM and 5 µM restored UPRmt-related protein levels. PA:OA (palmitic acid:oleic acid)-induced ER stress markers IRE1α (Inositol requiring enzyme-1), CHOP (C/EBP Homologous Protein), BIP (Binding Immunoglobulin Protein), and BAX (Bcl-2-associated X protein) were significantly reduced by ALA treatment. ALA also enhanced ER-mediated protein glycosylation and reduced oxidative stress, as evidenced by decreased GPX1 (Glutathione peroxidase 1), GSTP1 (glutathione S-transferase pi 1), and GSR (glutathione-disulfide reductase) expression and increased GSH (Glutathione) levels, and improved cellular senescence as shown by the markers ß-galactosidase, γH2Ax and Klotho-beta. CONCLUSIONS: In conclusion, ALA ameliorated ER stress, oxidative stress, and inflammation in HepG2 cells treated with palmitic and oleic acids, potentially offering therapeutic benefits for NAFLD providing a possible biochemical mechanism underlying ALA beneficial effects.


Subject(s)
Non-alcoholic Fatty Liver Disease , Thioctic Acid , Humans , Non-alcoholic Fatty Liver Disease/pathology , Thioctic Acid/pharmacology , Thioctic Acid/therapeutic use , Thioctic Acid/metabolism , Endoribonucleases/metabolism , Oleic Acid/pharmacology , Oleic Acid/metabolism , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response , Oxidative Stress , Endoplasmic Reticulum Stress , Hepatocytes/pathology , Cellular Senescence , Inflammation/pathology , Palmitic Acids/metabolism , Palmitic Acids/pharmacology , Liver/pathology , Palmitic Acid/pharmacology , Palmitic Acid/metabolism
8.
Pestic Biochem Physiol ; 198: 105725, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38225080

ABSTRACT

This study aimed to examine the effects of gibberellic acid (GBA) on growth, hemato-biochemical parameters related to liver functions, digestive enzymes, and immunological response in Oreochromis niloticus. Besides, the probable underlying mechanisms were explored by assessing antioxidant, apoptotic, and immune-related gene expression. Furthermore, the likelihood of restoration following alpha-lipoic acid (LIP) dietary supplementation was explored. The fish (average initial weight 30.75 ± 0.46) were equally classified into four groups: the control group, the LIP group (fed on a basal diet plus 600 mg/kg of LIP), the GBA group (exposed to 150 mg GBA/L), and the GBA + LIP group (exposed to 150 mg GBA/L and fed a diet containing LIP and GBA) for 60 days. The study findings showed that LIP supplementation significantly reduced GBA's harmful effects on survival rate, growth, feed intake, digestive enzymes, and antioxidant balance. Moreover, the GBA exposure significantly increased liver enzymes, stress markers, cholesterol, and triglyceride levels, all of which were effectively mitigated by the supplementation of LIP. Additionally, LIP addition to fish diets significantly minimized the histopathological alterations in the livers of GBA-treated fish, including fatty change, sharply clear cytoplasm with nuclear displacement to the cell periphery, single-cell necrosis, vascular congestion, and intralobular hemorrhages. The GBA-induced reduction in lysozyme activity, complement C3, and nitric oxide levels, together with the downregulation of antioxidant genes (cat and sod), was significantly restored by dietary LIP. Meanwhile, adding LIP to the GBA-exposed fish diets significantly corrected the aberrant expression of hsp70, caspase- 3, P53, pcna, tnf-a, and il-1ß in O. niloticus liver. Conclusively, dietary LIP supplementation could mitigate the harmful effects of GBA exposure on fish growth and performance, physiological conditions, innate immunity, antioxidant capability, inflammatory response, and cell apoptosis.


Subject(s)
Cichlids , Gibberellins , Thioctic Acid , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Dietary Supplements , Thioctic Acid/pharmacology , Thioctic Acid/metabolism , Cichlids/genetics , Oxidative Stress , Gene Expression
9.
Chem Biodivers ; 21(2): e202301371, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38069597

ABSTRACT

In the present study, a novel derivative, IOP-LA, was prepared by hybridizing antioxidant lipoic acid (LA) and our recently reported antioxidative marine phidianidine B-inspired indole/1,2,4-oxadiazole derivative. Our results demonstrated that IOP-LA could protect vascular endothelial cells (VECs) from oxidized low-density lipoprotein (oxLDL)-induced oxidative stress by activating the Nrf2 pathway, inhibit the production of atherosclerotic plaque, and promote the stability of atherosclerotic plaque in apoE-/- mice. Moreover, the protective effect of IOP-LA was superior to LA at the same concentration. Mechanistic studies revealed that IOP-LA significantly inhibited the increase of reactive oxygen species (ROS) levels and the translocation of nuclear factor kappa-B (NF-κB) nuclear induced by oxLDL through the nuclear factor erythroid2-related factor 2 (Nrf2) pathway. In summary, the data demonstrate that IOP-LA, as a new antioxidant, protects VECs from oxLDL-induced oxidative stress by activating the Nrf2 pathway. It is worth noting that this study provides a promising lead compound for the prevention and treatment of atherosclerosis.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Thioctic Acid , Animals , Mice , Thioctic Acid/pharmacology , Thioctic Acid/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Plaque, Atherosclerotic/metabolism , NF-E2-Related Factor 2/metabolism , Endothelial Cells , Atherosclerosis/drug therapy , Atherosclerosis/metabolism
10.
Toxicol Mech Methods ; 34(2): 176-188, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37904548

ABSTRACT

Imidacloprid (IMI), a widely used pesticide in agriculture and a potential food contaminant, poses significant health concerns. This study sought to comprehensively evaluate its neurotoxic effects while investigating the potential protective role of alpha-lipoic acid (ALA), a naturally occurring dietary antioxidant renowned for its capacity to combat oxidative stress, support cardiovascular health, and maintain optimal nerve function. In this study, 28 rats were divided evenly into four groups and administered oral treatments of corn oil, IMI, IMI + ALA, and ALA, respectively. The results of the study indicated that rats exposed to IMI exhibited significant neurobehavioral impairments, decreased levels of antioxidant enzymes and acetylcholinesterase activity, reduced expression of HO-1 and Nrf2, and increased levels of pro-inflammatory cytokines like IL-6 and TNF-α in their hippocampal tissues. Furthermore, histopathological analysis of the brain tissues, specifically cortex and hippocampus, from the IMI-treated group revealed varying degrees of neuronal degeneration. In contrast, rats co-administered ALA alongside IMI showed noticeable improvements in all the assessed toxicological parameters. This study underscores the vital significance of ALA as a potential therapeutic adjunct in mitigating the adverse neurobehavioral consequences of insecticide exposure. By harnessing the Nrf2/HO-1 pathway, ALA demonstrates its ability to shield against IMI-induced neurotoxicity, offering a promising avenue for enhancing public health and safety. As a result, our findings advocate for the incorporation of ALA as a daily dietary supplement to fortify resilience against oxidative stress-related neurobehavioral deficits linked to pesticide exposure, thereby advancing our understanding of neuroprotection strategies in the face of environmental challenges.


Subject(s)
Insecticides , Neonicotinoids , Nitro Compounds , Thioctic Acid , Rats , Animals , Thioctic Acid/pharmacology , Thioctic Acid/therapeutic use , Thioctic Acid/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , NF-E2-Related Factor 2/metabolism , Acetylcholinesterase/metabolism , Insecticides/toxicity , Oxidative Stress
11.
J Biol Chem ; 298(8): 102203, 2022 08.
Article in English | MEDLINE | ID: mdl-35764173

ABSTRACT

Lipoic acid is a sulfur-containing cofactor indispensable for the function of several metabolic enzymes. In microorganisms, lipoic acid can be salvaged from the surroundings by lipoate protein ligase A (LplA), an ATP-dependent enzyme. Alternatively, it can be synthesized by the sequential actions of lipoate protein ligase B (LipB) and lipoyl synthase (LipA). LipB takes up the octanoyl chain from C8-acyl carrier protein (C8-ACP), a byproduct of the type II fatty acid synthesis pathway, and transfers it to a conserved lysine of the lipoyl domain of a dehydrogenase. However, the molecular basis of its substrate recognition is still not fully understood. Using Escherichia coli LipB as a model enzyme, we show here that the octanoyl-transferase mainly recognizes the 4'-phosphopantetheine-tethered acyl-chain of its donor substrate and weakly binds the apo-acyl carrier protein. We demonstrate LipB can accept octanoate from its own ACP and noncognate ACPs, as well as C8-CoA. Furthermore, our 1H saturation transfer difference and 31P NMR studies demonstrate the binding of adenosine, as well as the phosphopantetheine arm of CoA to LipB, akin to binding to LplA. Finally, we show a conserved 71RGG73 loop, analogous to the lipoate-binding loop of LplA, is required for full LipB activity. Collectively, our studies highlight commonalities between LipB and LplA in their mechanism of substrate recognition. This knowledge could be of significance in the treatment of mitochondrial fatty acid synthesis related disorders.


Subject(s)
Acyltransferases/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Acyl Carrier Protein/metabolism , Acyltransferases/metabolism , Coenzyme A/metabolism , Escherichia coli/chemistry , Escherichia coli Proteins/metabolism , Ligases/metabolism , Pantetheine/analogs & derivatives , Thioctic Acid/metabolism
12.
Mol Microbiol ; 117(6): 1352-1365, 2022 06.
Article in English | MEDLINE | ID: mdl-35484915

ABSTRACT

Lipoic acid (LA) is a sulfur-containing cofactor covalently attached to key enzymes of central metabolism in prokaryotes and eukaryotes. LA can be acquired by scavenging, mediated by a lipoate ligase, or de novo synthesized by a pathway requiring an octanoyltransferase and a lipoate synthase. A more complex pathway, referred to as "lipoyl-relay", requires two additional proteins, GcvH, the glycine cleavage system H subunit, and an amidotransferase. This route was described so far in Bacillus subtilis and related Gram-positive bacteria, Saccharomyces cerevisiae, Homo sapiens, and Caenorhabditis elegans. Using collections of S. cerevisiae and B. subtilis mutants, defective in LA metabolism, we gathered evidence that allows us to propose for the first time that lipoyl-relay pathways are also present in parasitic protozoa. By a reverse genetic approach, we assigned octanoyltransferase and amidotransferase activity to the products of Tb927.11.9390 (TblipT) and Tb927.8.630 (TblipL) genes of Trypanosoma brucei, respectively. The B. subtilis model allowed us to identify the parasite amidotransferase as the target of lipoate analogs like 8-bromo-octanoic acid, explaining the complete loss of protein lipoylation and growth impairment caused by this compound in T. cruzi. This model could be instrumental for the screening of selective and more efficient chemotherapies against trypanosomiases.


Subject(s)
Metabolic Networks and Pathways , Thioctic Acid , Trypanosoma brucei brucei , Bacillus subtilis/metabolism , Ligases/metabolism , Metabolic Networks and Pathways/genetics , Saccharomyces cerevisiae/metabolism , Thioctic Acid/metabolism , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism
13.
Arch Biochem Biophys ; 750: 109811, 2023 12.
Article in English | MEDLINE | ID: mdl-37926405

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent liver disorder affecting a significant part of the global population. This study aimed to investigate the potential therapeutic effects of α-lipoic acid (α-LA) on the inflammatory response during simple steatosis development and progression into steatohepatitis. The study used the MASLD model in male Wistar rats that were fed a standard diet or a high-fat diet (HFD) for 8 weeks. Throughout the entire experiment, half of the animals received α-LA supplementation. The hepatic activity of pro-inflammatory n-6 and anti-inflammatory n-3 polyunsaturated fatty acid (PUFA) pathways and the concentration of arachidonic acid (AA) in selected lipid fractions were determined by the gas-liquid chromatography (GLC). The hepatic expression of proteins from inflammatory pathway was measured by the Western blot technique. The level of eicosanoids, cytokines and chemokines was assessed by the ELISA or multiplex assay kits. The results showed that α-LA supplementation attenuated the activity of n-6 PUFA pathway in FFA and DAG and increased the activity of n-3 PUFA pathway in PL, TAG and DAG. In addition, the administration of α-LA decreased the concentration of AA in DAG and FFA, indicating its potential protective effect on the deterioration of simple hepatic steatosis. The supplementation of α-LA also increased the expression of COX-1 and COX-2 with the lack of significant changes in prostaglandins profile. We observed an increase in the expression of 12/15-LOX, which was reflected in an increase in lipoxin A4 (LXA4) level. A decrease in pro-inflammatory cytokines and an increase in anti-inflammatory cytokines was also noticed in the liver of rats treated with HFD and α-LA. Our observations confirm that α-LA treatment has potential protective effects on inflammation development in the MASLD model. We believe that α-LA has a preventive impact when it comes to the progression of simple steatosis lesions to steatohepatitis.


Subject(s)
Fatty Liver , Thioctic Acid , Rats , Male , Animals , Thioctic Acid/pharmacology , Thioctic Acid/therapeutic use , Thioctic Acid/metabolism , Diet, High-Fat/adverse effects , Rats, Wistar , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/prevention & control , Liver/metabolism , Inflammation/drug therapy , Inflammation/prevention & control , Inflammation/metabolism , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism
14.
Cell Biochem Funct ; 41(8): 1403-1411, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37987234

ABSTRACT

Inflammation is a reaction of the immune system to infection and injury; in fact, it positioned at the center of metabolic disorders, particularly obesity, type 2 diabetes, and cardiovascular diseases. Thus play a major role not only in their development, but also exerts as a crucial linking factor among those diseases. In this regard, one of the strategies for tackling this problem is application of antioxidants to treat such diseases. The present study was performed to evaluate the synergistic effects of punicic acid (PUA) and alpha-lipoic acid (ALA) as antioxidants and radical scavenging reagents on the expression of some inflammatory and metabolism-related genes under oxidative stress in the muscle cells. The experimental treatments consisted of a range of 20, 40, 80, 160, and 320 µM of PUA, and 5, 25, 50, 100, and 200 µM of ALA with a 200 µM concentration of H2 O2 as an oxidative stress inducer. Accordingly, fatty acid treatments were applied for 24 h, and H2 O2 was treated for 1 h. Our results indicated that the simultaneous treatment of PUA and ALA at optimal concentrations (80 and 50 µM, respectively) decreased the expression of inflammation genes and increased the expression of regulatory genes (Pparγ, Pgc-1α) related to metabolism (p < .05). Unexpectedly, H2 O2 treatment increased the Fndc5 expression (p < .05). Maximal upregulation of Pparγ, Pgc-1α were obtained when fatty acids combination (PUA and ALA) were used in the culture of H2 O2 treated cells (p < .05). Therefore, our findings suggest that the simultaneous use of PUA and ALA fatty acids could reduce oxidative stress, and the expression of inflammatory genes, thereby improving the cell metabolism.


Subject(s)
Diabetes Mellitus, Type 2 , Thioctic Acid , Humans , Thioctic Acid/pharmacology , Thioctic Acid/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Antioxidants/pharmacology , Oxidative Stress , Linolenic Acids/pharmacology , Inflammation/drug therapy , Myoblasts/metabolism
15.
Graefes Arch Clin Exp Ophthalmol ; 261(3): 735-748, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36058948

ABSTRACT

PURPOSE: Oxidative stress and inflammation had been proved to play important role in the progression of diabetic keratopathy (DK). The excessive accumulation of AGEs and their bond to AGE receptor (RAGE) in corneas that cause the formation of oxygen radicals and the release of inflammatory cytokines, induce cell apoptosis. Our current study was aimed to evaluate the effect of ALA on AGEs accumulation as well as to study the molecular mechanism of ALA against AGE-RAGE axis mediated oxidative stress, apoptosis, and inflammation in HG-induced HCECs, so as to provide cytological basis for the treatment of DK. METHODS: HCECs were cultured in a variety concentration of glucose medium (5.5, 10, 25, 30, 40, and 50 mM) for 48 h. The cell proliferation was evaluated by CCK-8 assay. Apoptosis was investigated with the Annexin V- fluorescein isothiocyanate (V-FITC)/PI kit, while, the apoptotic cells were determined by flow cytometer and TUNEL cells apoptosis Kit. According to the results of cell proliferation and cell apoptosis, 25 mM glucose medium was used in the following HG experiment. The effect of ALA on HG-induced HCECs was evaluated. The HCECs were treated with 5.5 mM glucose (normal glucose group, NG group), 5.5 mM glucose + 22.5 mM mannitol (osmotic pressure control group, OP group), 25 mM glucose (high glucose group, HG group) and 25 mM glucose + ALA (HG + ALA group) for 24 and 48 h. The accumulation of intracellular AGEs was detected by ELISA kit. The RAGE, catalase (CAT), superoxide dismutase 2 (SOD2), cleaved cysteine-aspartic acid protease-3 (Cleaved caspase-3), Toll-like receptors 4 (TLR4), Nod-like receptor protein 3 (NLRP3) inflammasome, interleukin 1 beta (IL-1 ß), and interleukin 18 (IL-18) were quantified by RT-PCR, Western blotting, and Immunofluorescence, respectively. Reactive oxygen species (ROS) production was evaluated by fluorescence microscope and fluorescence microplate reader. RESULTS: When the glucose medium was higher than 25 mM, cell proliferation was significantly inhibited and apoptosis ratio was increased (P < 0.001). In HG environment, ALA treatment alleviated the inhibition of HCECs in a dose-dependent manner, 25 µM ALA was the minimum effective dose. ALA could significantly reduce the intracellular accumulation of AGEs (P < 0.001), activate protein and genes expression of CAT and SOD2 (P < 0.001), and therefore inhibited ROS-induced oxidative stress and cells apoptosis. Besides, ALA could effectively down-regulate the protein and gene level of RAGE, TLR4, NLRP3, IL-1B, IL-18 (P < 0.05), and therefore alleviated AGEs-RAGE-TLR4-NLRP3 pathway-induced inflammation in HG-induced HCECs. CONCLUSION: Our study indicated that ALA could be a desired treatment for DK due to its potential capacity of reducing accumulation of advanced glycation end products (AGEs) and down-regulating AGE-RAGE axis-mediated oxidative stress, cell apoptosis, and inflammation in high glucose (HG)-induced human corneal epithelial cells (HCECs), which may provide cytological basis for therapeutic targets that are ultimately of clinical benefit.


Subject(s)
Thioctic Acid , Humans , Thioctic Acid/pharmacology , Thioctic Acid/metabolism , Reactive Oxygen Species/metabolism , Interleukin-18/metabolism , Interleukin-18/pharmacology , Toll-Like Receptor 4/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , Inflammation/drug therapy , Apoptosis , Glucose/toxicity , Epithelial Cells/metabolism , Glycation End Products, Advanced/toxicity , Glycation End Products, Advanced/metabolism
16.
Biotechnol Lett ; 45(2): 273-286, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36586051

ABSTRACT

OBJECTIVE: To increase the production of (R)-α-lipoic acid directly from octanoic acid using engineered Escherichia coli with the regeneration of S-adenosylmethionine. RESULTS: The biosynthesis of (R)-α-lipoic acid (LA) in E. coli BL21(DE3) is improved by co-expression of lipoate-protein ligase A (LplA) from E. coli MG1655 and lipoate synthase (LipA) from Vibrio vulnificus. The engineered strain produces 20.99 µg l-1 of LA in shake flask cultures. The titers of LA are increased to 169.28 µg l-1 after the optimization of the medium components and fermentation conditions. We find that the [4Fe-4S] cluster is important for the activity of LipA and co-expression of iscSUA promotes the regeneration of the [4Fe-4S] cluster and leads to the highest LA titer of 589.30 µg l-1. CONCLUSION: The method described here can be widely applied for the biosynthesis of (R)-α-lipoic acid and other metabolites.


Subject(s)
Escherichia coli , Thioctic Acid , Escherichia coli/genetics , Escherichia coli/metabolism , Thioctic Acid/metabolism , Bacterial Proteins/genetics , Metabolic Engineering , Ligases
17.
Chem Biodivers ; 20(3): e202200928, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36650104

ABSTRACT

Smoking has been associated with NAFLD recently, thus might be a contributing factor for liver disease progression. In this study, we identified the modulative action of α-lipoic acid (α-LA), an organosulphur compound, towards heated tobacco product (HTP) and cigarette smoke extract (CSE)-induced oxidative stress and inflammation in human liver HepG2 cells. The cells were pre-treated with α-LA and exposed to tobacco extracts, and cytotoxicity, oxidative response (SOD, CAT activities and GSH, MDA levels), inflammation (nitrite, IL-6, AhR levels), and liver function (AST/ALT) were assessed. According to the results, a notable increase in oxidative response was observed with CSE, whereas GSH depletion and decreased SOD activity were the key toxicological events induced by HTP (p<0.05). The oxidative and inflammatory responses were ameliorated with α-LA treatment, particularly through GSH restoration and IL-6 modulation. To conclude, these findings on α-LA might contribute to the design of novel adjuvant therapies for people exposed to tobacco smoke.


Subject(s)
Thioctic Acid , Tobacco Products , Humans , Thioctic Acid/pharmacology , Thioctic Acid/metabolism , Nicotiana/metabolism , Interleukin-6/metabolism , Liver/metabolism , Oxidative Stress , Epithelial Cells/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Superoxide Dismutase/metabolism
18.
Int J Mol Sci ; 24(7)2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37047720

ABSTRACT

Protein probes, including ultrafiltrates from the placenta (UPla) and lung (ULu) of postnatal rabbits, were investigated in premature senescent HEK293 and HepG2 cells to explore whether they could modulate cellular senescence. Tris-Tricine-PAGE, gene ontology (GO), and LC-MS/MS analysis were applied to describe the characteristics of the ultrafiltrates. HEK293 and HepG2 cells (both under 25 passages) exposed to a sub-toxic concentration of hydrogen peroxide (H2O2, 300 µM) became senescent; UPla (10 µg/mL), ULu (10 µg/mL), as well as positive controls lipoic acid (10 µg/mL) and transferrin (10 µg/mL) were added along with H2O2 to the cells. Cell morphology; cellular proliferation; senescence-associated beta-galactosidase (SA-ß-X-gal) activity; expression of senescence biomarkers including p16 INK4A (p16), p21 Waf1/Cip1 (p21), HMGB1, MMP-3, TNF-α, IL-6, lamin B1, and phospho-histone H2A.X (γ-H2AX); senescence-related gene expression; reactive oxygen species (ROS) levels; and mitochondrial fission were examined. Tris-Tricine-PAGE revealed prominent detectable bands between 10 and 100 kDa. LC-MS/MS identified 150-180 proteins and peptides in the protein probes, and GO analysis demonstrated a distinct enrichment of proteins associated with "extracellular space" and "proteasome core complex". UPla and ULu modulated senescent cell morphology, improved cell proliferation, and decreased beta-galactosidase activity, intracellular and mitochondrial ROS production, and mitochondrial fission caused by H2O2. The results from this study demonstrated that UPla and Ulu, as well as lipoic acid and transferrin, could protect HEK293 and HepG2 cells from H2O2-induced oxidative damage via protecting mitochondrial homeostasis and thus have the potential to be explored in anti-aging therapies.


Subject(s)
Hydrogen Peroxide , Thioctic Acid , Animals , Humans , Rabbits , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Hep G2 Cells , Thioctic Acid/metabolism , Chromatography, Liquid , HEK293 Cells , Tandem Mass Spectrometry , Oxidative Stress , Cellular Senescence , beta-Galactosidase/metabolism , Transferrins/metabolism
19.
Int J Mol Sci ; 24(23)2023 Dec 04.
Article in English | MEDLINE | ID: mdl-38069431

ABSTRACT

Alpha-lipoic acid (ALA) is a natural antioxidant dithiol compound, exerting antiproliferative and antimetastatic effects in various cancer cell lines. In our study, we demonstrated that ALA reduces the cell growth of prostate cancer cells LNCaP and DU-145. Western blot results revealed that in both cancer cells, ALA, by upregulating pmTOR expression, reduced the protein content of two autophagy initiation markers, Beclin-1 and MAPLC3. Concomitantly, MTT assays showed that chloroquine (CQ) exposure, a well-known autophagy inhibitor, reduced cells' viability. This was more evident for treatment using the combination ALA + CQ, suggesting that ALA can reduce cells' viability by inhibiting autophagy. In addition, in DU-145 cells we observed that ALA affected the oxidative/redox balance system by deregulating the KEAP1/Nrf2/p62 signaling pathway. ALA decreased ROS production, SOD1 and GSTP1 protein expression, and significantly reduced the cytosolic and nuclear content of the transcription factor Nrf2, concomitantly downregulating p62, suggesting that ALA disrupted p62-Nrf2 feedback loop. Conversely, in LNCaP cells, ALA exposure upregulated both SOD1 and p62 protein expression, but did not affect the KEAP1/Nrf2/p62 signaling pathway. In addition, wound-healing, Western blot, and immunofluorescence assays evidenced that ALA significantly reduced the motility of LNCaP and DU-145 cells and downregulated the protein expression of TGFß1 and vimentin and the deposition of fibronectin. Finally, a soft agar assay revealed that ALA decreased the colony formation of both the prostate cancer cells by affecting the anchorage independent growth. Collectively, our in vitro evidence demonstrated that in prostate cancer cells, ALA reduces cell growth and counteracts both migration and invasion. Further studies are needed in order to achieve a better understanding of the underlined molecular mechanisms.


Subject(s)
Prostatic Neoplasms , Thioctic Acid , Male , Humans , Thioctic Acid/pharmacology , Thioctic Acid/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Superoxide Dismutase-1/metabolism , Cell Movement , Autophagy , Prostatic Neoplasms/drug therapy , Oxidative Stress
20.
Int J Mol Sci ; 24(19)2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37833939

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is characterized by the accumulation of lipids within hepatocytes, which compromises liver functionality following mitochondrial dysfunction and increased production of reactive oxygen species (ROS). Lipoic acid is one of the prosthetic groups of the pyruvate dehydrogenase complex also known for its ability to confer protection from oxidative damage because of its antioxidant properties. In this study, we aimed to investigate the effects of lipoic acid on lipotoxicity and mitochondrial dynamics in an in vitro model of liver steatosis. HepG2 cells were treated with palmitic acid and oleic acid (1:2) to induce steatosis, without and with 1 and 5 µM lipoic acid. Following treatments, cell proliferation and lipid droplets accumulation were evaluated. Mitochondrial functions were assessed through the evaluation of membrane potential, MitoTracker Red staining, expression of genes of the mitochondrial quality control, and analysis of energy metabolism by HPLC and Seahorse. We showed that lipoic acid treatment restored membrane potential to values comparable to control cells, as well as protected cells from mitochondrial fragmentation following PA:OA treatment. Furthermore, our data showed that lipoic acid was able to determine an increase in the expression of mitochondrial fusion genes and a decrease in mitochondrial fission genes, as well as to restore the bioenergetics of cells after treatment with palmitic acid and oleic acid. In conclusion, our data suggest that lipoic acid reduces lipotoxicity and improves mitochondrial functions in an in vitro model of steatosis, thus providing a potentially valuable pharmacological tool for NAFLD treatment.


Subject(s)
Non-alcoholic Fatty Liver Disease , Thioctic Acid , Humans , Thioctic Acid/pharmacology , Thioctic Acid/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Palmitic Acid/pharmacology , Palmitic Acid/metabolism , Oleic Acid/pharmacology , Oleic Acid/metabolism , Mitochondria/metabolism , Hepatocytes/metabolism , Oxidative Stress , Energy Metabolism , Liver/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL