Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.528
Filter
1.
Sci Adv ; 10(29): eadn4582, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39018392

ABSTRACT

The pyruvate dehydrogenase complex (PDHc) is a key megaenzyme linking glycolysis with the citric acid cycle. In mammalian PDHc, dihydrolipoamide acetyltransferase (E2) and the dihydrolipoamide dehydrogenase-binding protein (E3BP) form a 60-subunit core that associates with the peripheral subunits pyruvate dehydrogenase (E1) and dihydrolipoamide dehydrogenase (E3). The structure and stoichiometry of the fully assembled, mammalian PDHc or its core remained elusive. Here, we demonstrate that the human PDHc core is formed by 48 E2 copies that bind 48 E1 heterotetramers and 12 E3BP copies that bind 12 E3 homodimers. Cryo-electron microscopy, together with native and cross-linking mass spectrometry, confirmed a core model in which 8 E2 homotrimers and 12 E2-E2-E3BP heterotrimers assemble into a pseudoicosahedral particle such that the 12 E3BP molecules form six E3BP-E3BP intertrimer interfaces distributed tetrahedrally within the 60-subunit core. The even distribution of E3 subunits in the peripheral shell of PDHc guarantees maximum enzymatic activity of the megaenzyme.


Subject(s)
Cryoelectron Microscopy , Pyruvate Dehydrogenase Complex , Humans , Pyruvate Dehydrogenase Complex/metabolism , Pyruvate Dehydrogenase Complex/chemistry , Models, Molecular , Dihydrolipoamide Dehydrogenase/metabolism , Dihydrolipoamide Dehydrogenase/chemistry , Protein Multimerization , Protein Binding , Protein Subunits/metabolism , Protein Subunits/chemistry , Dihydrolipoyllysine-Residue Acetyltransferase/metabolism , Dihydrolipoyllysine-Residue Acetyltransferase/chemistry
2.
J Proteome Res ; 23(8): 3682-3695, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39037832

ABSTRACT

Dental caries is a chronic oral infectious disease, and Streptococcus mutans (S. mutans) plays an important role in the formation of dental caries. Trans-cinnamaldehyde (CA) exhibits broad-spectrum antibacterial activity; however, its target and mechanism of action of CA on S. mutans needs to be further explored. In this study, it was verified that CA could inhibit the growth and biofilm formation of S. mutans. Further proteomic analysis identified 33, 55, and 78 differentially expressed proteins (DEPs) in S. mutans treated with CA for 1, 2, and 4 h, respectively. Bioinformatics analysis showed that CA interfered with carbohydrate metabolism, glycolysis, pyruvate metabolism, and the TCA cycle, as well as amino acid metabolism of S. mutans. Protein interactions suggested that pyruvate dehydrogenase (PDH) plays an important role in the antibacterial effect of CA. Moreover, the upstream and downstream pathways related to PDH were verified by various assays, and the results proved that CA not only suppressed the glucose and sucrose consumption and inhibited glucosyltransferase (GTF) and lactate dehydrogenase (LDH) activities but also decreased the ATP production. Interestingly, the protein interaction, qRT-PCR, and molecular docking analysis showed that PDH might be the target of CA to fight S. mutans. In summary, the study shows that CA interferes with the carbohydrate metabolism of bacteria by inhibiting glycolysis and the tricarboxylic acid (TCA) cycle via binding to PDH, which verifies that PDH is a potential target for the development of new drugs against S. mutans.


Subject(s)
Acrolein , Carbohydrate Metabolism , Molecular Docking Simulation , Pyruvate Dehydrogenase Complex , Streptococcus mutans , Streptococcus mutans/drug effects , Streptococcus mutans/genetics , Streptococcus mutans/enzymology , Acrolein/pharmacology , Acrolein/analogs & derivatives , Acrolein/metabolism , Carbohydrate Metabolism/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Pyruvate Dehydrogenase Complex/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Glycolysis/drug effects , Biofilms/drug effects , Biofilms/growth & development , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/antagonists & inhibitors , Proteomics/methods , Dental Caries/microbiology , Citric Acid Cycle/drug effects , Adenosine Triphosphate/metabolism
3.
Sci Adv ; 10(30): eado2825, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39058774

ABSTRACT

Ethylene plays its essential roles in plant development, growth, and defense responses by controlling the transcriptional reprograming, in which EIN2-C-directed regulation of histone acetylation is the first key step for chromatin to perceive ethylene signaling. But how the nuclear acetyl coenzyme A (acetyl CoA) is produced to ensure the ethylene-mediated histone acetylation is unknown. Here we report that ethylene triggers the accumulation of the pyruvate dehydrogenase complex (PDC) in the nucleus to synthesize nuclear acetyl CoA to regulate ethylene response. PDC is identified as an EIN2-C nuclear partner, and ethylene triggers its nuclear accumulation. Mutations in PDC lead to an ethylene hyposensitivity that results from the reduction of histone acetylation and transcription activation. Enzymatically active nuclear PDC synthesizes nuclear acetyl CoA for EIN2-C-directed histone acetylation and transcription regulation. These findings uncover a mechanism by which PDC-EIN2 converges the mitochondrial enzyme-mediated nuclear acetyl CoA synthesis with epigenetic and transcriptional regulation for plant hormone response.


Subject(s)
Acetyl Coenzyme A , Arabidopsis Proteins , Arabidopsis , Cell Nucleus , Ethylenes , Gene Expression Regulation, Plant , Histones , Pyruvate Dehydrogenase Complex , Acetylation , Ethylenes/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Pyruvate Dehydrogenase Complex/genetics , Histones/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Cell Nucleus/metabolism , Acetyl Coenzyme A/metabolism , Transcription, Genetic , Mutation , Signal Transduction , Receptors, Cell Surface
4.
Microb Biotechnol ; 17(6): e14514, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923400

ABSTRACT

Pyruvate dehydrogenase (PDH) catalyses the irreversible decarboxylation of pyruvate to acetyl-CoA, which feeds the tricarboxylic acid cycle. We investigated how the loss of PDH affects metabolism in Pseudomonas putida. PDH inactivation resulted in a strain unable to utilize compounds whose assimilation converges at pyruvate, including sugars and several amino acids, whereas compounds that generate acetyl-CoA supported growth. PDH inactivation also resulted in the loss of carbon catabolite repression (CCR), which inhibits the assimilation of non-preferred compounds in the presence of other preferred compounds. Pseudomonas putida can degrade many aromatic compounds, most of which produce acetyl-CoA, making it useful for biotransformation and bioremediation. However, the genes involved in these metabolic pathways are often inhibited by CCR when glucose or amino acids are also present. Our results demonstrate that the PDH-null strain can efficiently degrade aromatic compounds even in the presence of other preferred substrates, which the wild-type strain does inefficiently, or not at all. As the loss of PDH limits the assimilation of many sugars and amino acids and relieves the CCR, the PDH-null strain could be useful in biotransformation or bioremediation processes that require growth with mixtures of preferred substrates and aromatic compounds.


Subject(s)
Catabolite Repression , Pseudomonas putida , Pyruvate Dehydrogenase Complex , Pseudomonas putida/genetics , Pseudomonas putida/metabolism , Pseudomonas putida/enzymology , Pyruvate Dehydrogenase Complex/metabolism , Pyruvate Dehydrogenase Complex/genetics , Hydrocarbons, Aromatic/metabolism , Biodegradation, Environmental , Acetyl Coenzyme A/metabolism , Pyruvic Acid/metabolism , Gene Deletion , Metabolic Networks and Pathways/genetics
5.
J Endocrinol ; 262(2)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38860519

ABSTRACT

Liraglutide, a glucagon-like peptide-1 receptor (GLP-1R) agonist used for the treatment of T2D, has been shown to alleviate diabetic cardiomyopathy (DbCM) in experimental T2D, which was associated with increased myocardial glucose oxidation. To determine whether this increase in glucose oxidation is necessary for cardioprotection, we hypothesized that liraglutide's ability to alleviate DbCM would be abolished in mice with cardiomyocyte-specific deletion of pyruvate dehydrogenase (PDH; Pdha1CM-/- mice), the rate-limiting enzyme of glucose oxidation. Male Pdha1CM-/- mice and their α-myosin heavy chain Cre expressing littermates (αMHCCre mice) were subjected to experimental T2D via 10 weeks of high-fat diet supplementation, with a single low-dose injection of streptozotocin (75 mg/kg) provided at week 4. All mice were randomized to treatment with either vehicle control or liraglutide (30 µg/kg) twice daily during the final 2.5 weeks, with cardiac function assessed via ultrasound echocardiography. As expected, liraglutide treatment improved glucose homeostasis in both αMHCCre and Pdha1CM-/- mice with T2D, in the presence of mild weight loss. Parameters of systolic function were unaffected by liraglutide treatment in both αMHCCre and Pdha1CM-/- mice with T2D. However, liraglutide treatment alleviated diastolic dysfunction in αMHCCre mice, as indicated by an increase and decrease in the e'/a' and E/e' ratios, respectively. Conversely, liraglutide failed to rescue these indices of diastolic dysfunction in Pdha1CM-/- mice. Our findings suggest that increases in glucose oxidation are necessary for GLP-1R agonist mediated alleviation of DbCM. As such, strategies aimed at increasing PDH activity may represent a novel approach for the treatment of DbCM.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Liraglutide , Animals , Liraglutide/pharmacology , Liraglutide/therapeutic use , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/metabolism , Male , Mice , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Mice, Knockout , Pyruvate Dehydrogenase Complex/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Blood Glucose/metabolism , Blood Glucose/drug effects , Mice, Inbred C57BL , Diet, High-Fat , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose/metabolism
6.
Neurochem Int ; 178: 105791, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38880231

ABSTRACT

Long-term studies have confirmed a causal relationship between the development of neurodegenerative processes and vitamin B1 (thiamine) deficiency. However, the biochemical mechanisms underlying the high neurotropic activity of thiamine are not fully understood. At the same time, there is increasing evidence that vitamin B1, in addition to its coenzyme functions, may have non-coenzyme activities that are particularly important for neurons. To elucidate which effects of vitamin B1 in neurons are due to its coenzyme function and which are due to its non-coenzyme activity, we conducted a comparative study of the effects of thiamine and its derivative, 3-decyloxycarbonylmethyl-5-(2-hydroxyethyl)-4-methyl-1,3-thiazolium chloride (DMHT), on selected processes in synaptosomes. The ability of DMHT to effectively compete with thiamine for binding to thiamine-binding sites on the plasma membrane of synaptosomes and to participate as a substrate in the thiamine pyrophosphokinase reaction was demonstrated. In experiments with rat brain synaptosomes, unidirectional effects of DMHT and thiamine on the activity of the pyruvate dehydrogenase complex (PDC) and on the incorporation of radiolabeled [2-14C]pyruvate into acetylcholine were demonstrated. The observed effects of thiamine and DMHT on the modulation of acetylcholine synthesis can be explained by suggesting that both compounds, which interact in cells with enzymes of thiamine metabolism, are phosphorylated and exert an inhibitory/activating effect (concentration-dependent) on PDC activity by affecting the regulatory enzymes of the complex. Such effects were not observed in the presence of structural analogues of thiamine and DMHT without a 2-hydroxyethyl substituent at position 5 of the thiazolium cycle. The effect of DMHT on the plasma membrane Ca-ATPase was similar to that of thiamine. At the same time, DMHT showed high cytostatic activity against neuroblastoma cells.


Subject(s)
Rats, Wistar , Synaptosomes , Thiamine , Animals , Synaptosomes/metabolism , Synaptosomes/drug effects , Rats , Thiamine/pharmacology , Thiamine/metabolism , Male , Acetylcholine/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Thiazoles/pharmacology , Coenzymes/metabolism , Brain/metabolism , Brain/drug effects , Pyruvic Acid/metabolism , Pyruvic Acid/pharmacology
7.
J Biol Chem ; 300(7): 107412, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38796064

ABSTRACT

The heart alters the rate and relative oxidation of fatty acids and glucose based on availability and energetic demand. Insulin plays a crucial role in this process diminishing fatty acid and increasing glucose oxidation when glucose availability increases. Loss of insulin sensitivity and metabolic flexibility can result in cardiovascular disease. It is therefore important to identify mechanisms by which insulin regulates substrate utilization in the heart. Mitochondrial pyruvate dehydrogenase (PDH) is the key regulatory site for the oxidation of glucose for ATP production. Nevertheless, the impact of insulin on PDH activity has not been fully delineated, particularly in the heart. We sought in vivo evidence that insulin stimulates cardiac PDH and that this process is driven by the inhibition of fatty acid oxidation. Mice injected with insulin exhibited dephosphorylation and activation of cardiac PDH. This was accompanied by an increase in the content of malonyl-CoA, an inhibitor of carnitine palmitoyltransferase 1 (CPT1), and, thus, mitochondrial import of fatty acids. Administration of the CPT1 inhibitor oxfenicine was sufficient to activate PDH. Malonyl-CoA is produced by acetyl-CoA carboxylase (ACC). Pharmacologic inhibition or knockout of cardiac ACC diminished insulin-dependent production of malonyl-CoA and activation of PDH. Finally, circulating insulin and cardiac glucose utilization exhibit daily rhythms reflective of nutritional status. We demonstrate that time-of-day-dependent changes in PDH activity are mediated, in part, by ACC-dependent production of malonyl-CoA. Thus, by inhibiting fatty acid oxidation, insulin reciprocally activates PDH. These studies identify potential molecular targets to promote cardiac glucose oxidation and treat heart disease.


Subject(s)
Fatty Acids , Insulin , Myocardium , Oxidation-Reduction , Pyruvate Dehydrogenase Complex , Animals , Insulin/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Mice , Myocardium/metabolism , Myocardium/enzymology , Fatty Acids/metabolism , Acetyl-CoA Carboxylase/metabolism , Acetyl-CoA Carboxylase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Malonyl Coenzyme A/metabolism , Male , Mice, Knockout , Glucose/metabolism , Mice, Inbred C57BL
8.
Diabetes ; 73(7): 1072-1083, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38608261

ABSTRACT

Insulin resistance is a risk factor for type 2 diabetes, and exercise can improve insulin sensitivity. However, following exercise, high circulating fatty acid (FA) levels might counteract this. We hypothesized that such inhibition would be reduced by forcibly increasing carbohydrate oxidation through pharmacological activation of the pyruvate dehydrogenase complex (PDC). Insulin-stimulated glucose uptake was examined with a crossover design in healthy young men (n = 8) in a previously exercised and a rested leg during a hyperinsulinemic-euglycemic clamp 5 h after one-legged exercise with 1) infusion of saline, 2) infusion of intralipid imitating circulating FA levels during recovery from whole-body exercise, and 3) infusion of intralipid + oral PDC activator, dichloroacetate (DCA). Intralipid infusion reduced insulin-stimulated glucose uptake by 19% in the previously exercised leg, which was not observed in the contralateral rested leg. Interestingly, this effect of intralipid in the exercised leg was abolished by DCA, which increased muscle PDC activity (130%) and flux (acetylcarnitine 130%) and decreased inhibitory phosphorylation of PDC on Ser293 (∼40%) and Ser300 (∼80%). Novel insight is provided into the regulatory interaction between glucose and lipid metabolism during exercise recovery. Coupling exercise and PDC flux activation upregulated the capacity for both glucose transport (exercise) and oxidation (DCA), which seems necessary to fully stimulate insulin-stimulated glucose uptake during recovery.


Subject(s)
Exercise , Insulin , Muscle, Skeletal , Pyruvate Dehydrogenase Complex , Humans , Male , Exercise/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Insulin/metabolism , Insulin/blood , Pyruvate Dehydrogenase Complex/metabolism , Adult , Young Adult , Glucose Clamp Technique , Cross-Over Studies , Dichloroacetic Acid/pharmacology , Insulin Resistance/physiology , Fatty Acids/metabolism , Glucose/metabolism , Soybean Oil/pharmacology , Post-Exercise Recovery , Emulsions , Phospholipids
9.
Nature ; 629(8010): 184-192, 2024 May.
Article in English | MEDLINE | ID: mdl-38600378

ABSTRACT

Glucocorticoids represent the mainstay of therapy for a broad spectrum of immune-mediated inflammatory diseases. However, the molecular mechanisms underlying their anti-inflammatory mode of action have remained incompletely understood1. Here we show that the anti-inflammatory properties of glucocorticoids involve reprogramming of the mitochondrial metabolism of macrophages, resulting in increased and sustained production of the anti-inflammatory metabolite itaconate and consequent inhibition of the inflammatory response. The glucocorticoid receptor interacts with parts of the pyruvate dehydrogenase complex whereby glucocorticoids provoke an increase in activity and enable an accelerated and paradoxical flux of the tricarboxylic acid (TCA) cycle in otherwise pro-inflammatory macrophages. This glucocorticoid-mediated rewiring of mitochondrial metabolism potentiates TCA-cycle-dependent production of itaconate throughout the inflammatory response, thereby interfering with the production of pro-inflammatory cytokines. By contrast, artificial blocking of the TCA cycle or genetic deficiency in aconitate decarboxylase 1, the rate-limiting enzyme of itaconate synthesis, interferes with the anti-inflammatory effects of glucocorticoids and, accordingly, abrogates their beneficial effects during a diverse range of preclinical models of immune-mediated inflammatory diseases. Our findings provide important insights into the anti-inflammatory properties of glucocorticoids and have substantial implications for the design of new classes of anti-inflammatory drugs.


Subject(s)
Anti-Inflammatory Agents , Glucocorticoids , Inflammation , Macrophages , Mitochondria , Succinates , Animals , Female , Humans , Male , Mice , Anti-Inflammatory Agents/pharmacology , Carboxy-Lyases/metabolism , Carboxy-Lyases/antagonists & inhibitors , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Cytokines/immunology , Cytokines/metabolism , Glucocorticoids/pharmacology , Glucocorticoids/metabolism , Hydro-Lyases/deficiency , Hydro-Lyases/genetics , Inflammation/drug therapy , Inflammation/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Receptors, Glucocorticoid/metabolism , Succinates/metabolism , Enzyme Activation/drug effects
10.
Meat Sci ; 213: 109510, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38598967

ABSTRACT

This research aimed to explore the potential influence of mitochondria on the rate of anaerobic glycolysis. We hypothesized that mitochondria could reduce the rate of anaerobic glycolysis and pH decline by metabolizing a portion of glycolytic pyruvate. We utilized an in vitro model and incorporated CPI-613 and Avidin to inhibit pyruvate dehydrogenase (PDH) and pyruvate carboxylase (PC), respectively. Four treatments were tested: 400 µM CPI-613, 1.5 U/ml Avidin, 400 µM CPI-613 + 1.5 U/ml Avidin, or control. Glycolytic metabolites and pH of the in vitro model were evaluated throughout a 1440-min incubation period. CPI-613-containing treatments, with or without Avidin, decreased pH levels and increased glycogen degradation and lactate accumulation compared to the control and Avidin treatments (P < 0.05), indicating increased glycolytic flux. In a different experiment, two treatments, 400 µM CPI-613 or control, were employed to track the fates of pyruvate using [13C6]glucose. CPI-613 reduced the contribution of glucose carbon to tricarboxylic acid cycle intermediates compared to control (P < 0.05). To test whether the acceleration of acidification in reactions containing CPI-613 was due to an increase in the activity of key enzymes of glycogenolysis and glycolysis, we evaluated the activities of glycogen phosphorylase, phosphofructokinase, and pyruvate kinase in the presence or absence of 400 µM CPI-613. The CPI-613 treatment did not elicit an alteration in the activity of these three enzymes. These findings indicate that inhibiting PDH increases the rate of anaerobic glycolysis and pH decline, suggesting that mitochondria are potential regulators of postmortem metabolism.


Subject(s)
Glycogen , Glycolysis , Pyruvate Dehydrogenase Complex , Animals , Anaerobiosis , Glucose/metabolism , Glycogen/metabolism , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Mitochondria/metabolism , Postmortem Changes , Pyruvate Carboxylase/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Pyruvic Acid/metabolism , Swine
11.
Resuscitation ; 198: 110160, 2024 May.
Article in English | MEDLINE | ID: mdl-38428722

ABSTRACT

INTRODUCTION: Elevated lactate is associated with mortality after cardiac arrest. Thiamine, a cofactor of pyruvate dehydrogenase, is necessary for aerobic metabolism. In a mouse model of cardiac arrest, thiamine improved pyruvate dehydrogenase activity, survival and neurologic outcome. AIM: To determine if thiamine would decrease lactate and increase oxygen consumption after in-hospital cardiac arrest. METHODS: Randomized, double-blind, placebo-controlled phase II trial. Adult patients with arrest within 12 hours, mechanically ventilated, with lactate ≥ 3 mmol/L were included. Randomization was stratified by lactate > 5 or ≤ 5 mmol/L. Thiamine 500 mg or placebo was administered every 12 hours for 3 days. The primary outcome of lactate was checked at baseline, 6, 12, 24, and 48 hours, and compared using a linear mixed model, accounting for repeated measures. Secondary outcomes included oxygen consumption, pyruvate dehydrogenase, and mortality. RESULTS: Enrollments stopped after 36 patients due Data Safety and Monitoring Board concern about potential harm in an unplanned subgroup analysis. There was no overall difference in lactate (mean difference at 48 hours 1.5 mmol/L [95% CI -3.1-6.1], global p = 0.88) or any secondary outcomes. In those with randomization lactate > 5 mmol/L, mortality was 92% (11/12) with thiamine and 67% (8/12) with placebo (p = 0.32). In those with randomization lactate ≤ 5 mmol/L mortality was 17% (1/6) with thiamine and 67% (4/6) with placebo (p = 0.24). There was a significant interaction between randomization lactate and the effect of thiamine on survival (p = 0.03). CONCLUSIONS: In this single center trial thiamine had no overall effect on lactate after in-hospital cardiac arrest.


Subject(s)
Heart Arrest , Thiamine , Humans , Thiamine/therapeutic use , Thiamine/administration & dosage , Male , Double-Blind Method , Female , Middle Aged , Heart Arrest/therapy , Heart Arrest/mortality , Aged , Lactic Acid/blood , Oxygen Consumption/drug effects , Cardiopulmonary Resuscitation/methods , Vitamin B Complex/therapeutic use , Vitamin B Complex/administration & dosage , Pyruvate Dehydrogenase Complex/metabolism
12.
Science ; 383(6690): 1484-1492, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38547260

ABSTRACT

Cellular purines, particularly adenosine 5'-triphosphate (ATP), fuel many metabolic reactions, but less is known about the direct effects of pyrimidines on cellular metabolism. We found that pyrimidines, but not purines, maintain pyruvate oxidation and the tricarboxylic citric acid (TCA) cycle by regulating pyruvate dehydrogenase (PDH) activity. PDH activity requires sufficient substrates and cofactors, including thiamine pyrophosphate (TPP). Depletion of cellular pyrimidines decreased TPP synthesis, a reaction carried out by TPP kinase 1 (TPK1), which reportedly uses ATP to phosphorylate thiamine (vitamin B1). We found that uridine 5'-triphosphate (UTP) acts as the preferred substrate for TPK1, enabling cellular TPP synthesis, PDH activity, TCA-cycle activity, lipogenesis, and adipocyte differentiation. Thus, UTP is required for vitamin B1 utilization to maintain pyruvate oxidation and lipogenesis.


Subject(s)
Citric Acid Cycle , Lipogenesis , Pyrimidines , Pyruvate Dehydrogenase Complex , Pyruvates , Adenosine Triphosphate/metabolism , Pyrimidines/metabolism , Pyruvates/metabolism , Thiamine/metabolism , Thiamine Pyrophosphate/metabolism , Uridine Triphosphate/metabolism , Oxidation-Reduction , Protein Kinases/metabolism , Humans , HeLa Cells , Pyruvate Dehydrogenase Complex/metabolism
13.
Sci Rep ; 14(1): 5665, 2024 03 07.
Article in English | MEDLINE | ID: mdl-38453992

ABSTRACT

Pyruvate dehydrogenase kinases (PDKs) play a key role in glucose metabolism by exerting negative regulation over pyruvate dehyrogenase complex (PDC) activity through phosphorylation. Inhibition of PDKs holds the potential to enhance PDC activity, prompting cells to adopt a more aerobic metabolic profile. Consequently, PDKs emerge as promising targets for condition rooted in metabolic dysregulation, including malignance and diabetes. However, a comprehensive exploration of the distinct contribution of various PDK family members, particularly PDK3, across diverse tumor types remain incomplete. This study undertakes a systematic investigation of PDK family expression patterns, forging association with clinical parameters, using data from the TCGA and GTEx datasets. Survival analysis of PDKs is executed through both Kaplan-Meier analysis and COX regression analysis. Furthermore, the extent of immune infiltration is assessed by leveraging the CIBERSORT algorithm. Our study uncovers pronounced genetic heterogeneity among PDK family members, coupled with discernible clinical characteristic. Significantly, the study establishes the potential utility of PDK family genes as prognostic indicators and as predictors of therapeutic response. Additionally, our study sheds light on the immune infiltration profile of PDK family. The results showed the intimate involvement of these genes in immune-related metrics, including immune scoring, immune subtypes, tumor-infiltrating lymphocytes, and immune checkpoints expression. In sum, the findings of this study offer insightful strategies to guide the therapeutic direction, aiming at leveraging the impact of PDK family genes in cancer treatment.


Subject(s)
Neoplasms , Protein Serine-Threonine Kinases , Humans , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Neoplasms/metabolism , Prognosis , Pyruvates , Pyruvate Dehydrogenase Complex/metabolism
14.
Bioorg Chem ; 144: 107160, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301426

ABSTRACT

Metabolism is reprogrammed in a variety of cancer cells to ensure their rapid proliferation. Cancer cells prefer to utilize glycolysis to produce energy as well as to provide large amounts of precursors for their division. In this process, cancer cells inhibit the activity of pyruvate dehydrogenase complex (PDC) by upregulating the expression of pyruvate dehydrogenase kinases (PDKs). Inhibiting the activity of PDKs in cancer cells can effectively block this metabolic transition in cancer cells, while also activating mitochondrial oxidative metabolism and promoting apoptosis of cancer cells. To this day, the study of PDKs inhibitors has become one of the research hotspots in the field of medicinal chemistry. Novel structures targeting PDKs are constantly being discovered, and some inhibitors have entered the clinical research stage. Here, we reviewed the research progress of PDKs inhibitors in recent years and classified them according to the PDKs binding sites they acted on, aiming to summarize the structural characteristics of inhibitors acting on different binding sites and explore their clinical application value. Finally, the shortcomings of some PDKs inhibitors and the further development direction of PDKs inhibitors are discussed.


Subject(s)
Protein Serine-Threonine Kinases , Pyruvate Dehydrogenase Complex , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Glycolysis , Binding Sites
15.
Int J Biol Macromol ; 262(Pt 1): 129970, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38325689

ABSTRACT

In humans and animals, the pyruvate dehydrogenase kinase (PDK) family proteins (PDKs 1-4) are excessively activated in metabolic disorders such as obesity, diabetes, and cancer, inhibiting the activity of pyruvate dehydrogenase (PDH) which plays a crucial role in energy and fatty acid metabolism and impairing its function. Intervention and regulation of PDH activity have become important research approaches for the treatment of various metabolic disorders. In this study, a small molecule (g25) targeting PDKs and activating PDH, was identified through multi-level computational screening methods. In vivo and in vitro experiments have shown that g25 activated the activity of PDH and reduced plasma lactate and triglyceride level. Besides, g25 significantly decreased hepatic fat deposition in a diet-induced obesity mouse model. Furthermore, g25 enhanced the tumor-inhibiting activity of cisplatin when used in combination. Molecular dynamics simulations and in vitro kinase assay also revealed the specificity of g25 towards PDK2. Overall, these findings emphasize the importance of targeting the PDK/PDH axis to regulate PDH enzyme activity in the treatment of metabolic disorders, providing directions for future related research. This study provides a possible lead compound for the PDK/PDH axis related diseases and offers insights into the regulatory mechanisms of this pathway in diseases.


Subject(s)
Metabolic Diseases , Neoplasms , Animals , Mice , Humans , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Phosphorylation , Metabolic Diseases/drug therapy , Obesity
16.
Sci Adv ; 10(6): eadj6358, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38324697

ABSTRACT

The Escherichia coli pyruvate dehydrogenase complex (PDHc) is a ~5 MDa assembly of the catalytic subunits pyruvate dehydrogenase (E1), dihydrolipoamide acetyltransferase (E2), and dihydrolipoamide dehydrogenase (E3). The PDHc core is a cubic complex of eight E2 homotrimers. Homodimers of the peripheral subunits E1 and E3 associate with the core by binding to the peripheral subunit binding domain (PSBD) of E2. Previous reports indicated that 12 E1 dimers and 6 E3 dimers bind to the 24-meric E2 core. Using an assembly arrested E2 homotrimer (E23), we show that two of the three PSBDs in the E23 dimerize, that each PSBD dimer cooperatively binds two E1 dimers, and that E3 dimers only bind to the unpaired PSBD in E23. This mechanism is preserved in wild-type PDHc, with an E1 dimer:E2 monomer:E3 dimer stoichiometry of 16:24:8. The conserved PSBD dimer interface indicates that PSBD dimerization is the previously unrecognized architectural determinant of gammaproteobacterial PDHc megacomplexes.


Subject(s)
Dihydrolipoamide Dehydrogenase , Dihydrolipoyllysine-Residue Acetyltransferase , Escherichia coli , Pyruvate Dehydrogenase Complex , Dihydrolipoamide Dehydrogenase/genetics , Dihydrolipoamide Dehydrogenase/chemistry , Dihydrolipoamide Dehydrogenase/metabolism , Dihydrolipoyllysine-Residue Acetyltransferase/chemistry , Dihydrolipoyllysine-Residue Acetyltransferase/metabolism , Dimerization , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Pyruvate Dehydrogenase Complex/chemistry , Pyruvate Dehydrogenase Complex/metabolism
17.
FEBS J ; 291(1): 132-141, 2024 01.
Article in English | MEDLINE | ID: mdl-37789611

ABSTRACT

In the present study, cryo-electron tomography was used to investigate the localization of 2-oxoacid dehydrogenase complexes (OADCs) in cardiac mitochondria and mitochondrial inner membrane samples. Two classes of ordered OADC inner cores with different symmetries were distinguished and their quaternary structures modeled. One class corresponds to pyruvate dehydrogenase complexes and the other to dehydrogenase complexes of α-ketoglutarate and branched-chain α-ketoacids. OADCs were shown to be localized in close proximity to membrane-embedded respirasomes, as observed both in densely packed lamellar cristae of cardiac mitochondria and in ruptured mitochondrial samples where the dense packing is absent. This suggests the specificity of the OADC-respirasome interaction, which allows localized NADH/NAD+ exchange between OADCs and complex I of the respiratory chain. The importance of this local coupling is based on OADCs being the link between respiration, glycolysis and amino acid metabolism. The coupling of these basic metabolic processes can vary in different tissues and conditions and may be involved in the development of various pathologies. The present study shows that this important and previously missing parameter of mitochondrial complex coupling can be successfully assessed using cryo-electron tomography.


Subject(s)
Keto Acids , Pyruvate Dehydrogenase Complex , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide) , Pyruvate Dehydrogenase Complex/metabolism , Mitochondria, Heart/metabolism , Ketoglutaric Acids , Ketoglutarate Dehydrogenase Complex/metabolism
18.
Acta Biochim Biophys Sin (Shanghai) ; 56(1): 44-53, 2024 01 25.
Article in English | MEDLINE | ID: mdl-37905340

ABSTRACT

The incidence and related death of hepatocellular carcinoma (HCC) have increased over the past decades. However, the molecular mechanisms underlying HCC pathogenesis are not fully understood. Long noncoding RNA (lncRNA) RP11-495P10.1 has been proven to be closely associated with the progression of prostate cancer, but its role and specific mechanism in HCC are still unknown. Here, we identify that RP11-495P10.1 is highly expressed in HCC tissues and cells and contributes to the proliferation of HCC cells. Moreover, this study demonstrates that RP11-495P10.1 affects the proliferation of HCC by negatively regulating the expression of nuclear receptor subfamily 4 group a member 3 (NR4A3). Glycometabolism reprogramming is one of the main characteristics of tumor cells. In this study, we discover that RP11-495P10.1 regulates glycometabolism reprogramming by changing the expression of pyruvate dehydrogenase kinase 1 (PDK1) and pyruvate dehydrogenase (PDH), thus contributing to the proliferation of HCC cells. Furthermore, knockdown of RP11-495P10.1 increases enrichment of H3K27Ac in the promoter of NR4A3 by promoting the activity of PDH and the production of acetyl-CoA, which leads to the increased transcription of NR4A3. Altogether, RP11-495P10.1 promotes HCC cell proliferation by regulating the reprogramming of glucose metabolism and acetylation of the NR4A3 promoter via the PDK1/PDH axis, which provides an lncRNA-oriented therapeutic strategy for the diagnosis and treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , RNA, Long Noncoding , Receptors, Steroid , Humans , Male , Acetylation , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Glucose , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Complex/metabolism
19.
Bioorg Med Chem Lett ; 98: 129571, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38036274

ABSTRACT

Pyruvate dehydrogenase complex (PDHc) is suppressed in some cancer types but overexpressed in others. To understand its contrasting oncogenic roles, there is a need for selective PDHc inhibitors. Its E1-subunit (PDH E1) is a thiamine pyrophosphate (TPP)-dependent enzyme and catalyses the first and rate-limiting step of the complex. In a recent study, we reported a series of ester-based thiamine analogues as selective TPP-competitive PDH E1 inhibitors with low nanomolar affinity. However, when the ester linker was replaced with an amide for stability reasons, the binding affinity was significantly reduced. In this study, we show that an amino-oxetane bioisostere of the amide improves the affinity and maintains stability towards esterase-catalysed hydrolysis.


Subject(s)
Pyruvate Dehydrogenase Complex , Thiamine Pyrophosphate , Thiamine , Amides , Esters , Oxidoreductases , Pyruvate Dehydrogenase Complex/antagonists & inhibitors , Pyruvate Dehydrogenase Complex/metabolism , Pyruvates , Thiamine/pharmacology , Thiamine Pyrophosphate/metabolism , Thiamine Pyrophosphate/pharmacology
20.
Eur J Med Chem ; 264: 116008, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38056298

ABSTRACT

Proliferating cancer cells are characterized by the Warburg effect, a metabolic alteration in which ATP is generated from cytoplasmic glycolysis instead of oxidative phosphorylation. The pyruvate dehydrogenase complex/pyruvate dehydrogenase kinase (PDC/PDK) axis plays a crucial role in this effect and has been identified as a potential target for anticancer drug development. Herein, we present the discovery and pharmacological evaluation of potent PDK inhibitors targeting the PDK/PDC axis. We successfully identified 6 compounds from a small molecule library through a structure-based virtual screening campaign and evaluated their enzymatic inhibitory potencies for PDK1-4. Our results indicated that compound 1 exhibited submicromolar inhibitory activities against PDK1-3 (IC50 = 109.3, 135.8, and 458.7 nM, respectively), but is insensitive to PDK4 (IC50 = 8.67 µM). Furthermore, compound 1 inhibited the proliferation of A549 cells with an EC50 value of 10.7 µM. In addition, compound 1 induced cell apoptosis, arrested the cell cycle at the S phase, and reduced cell invasion and migration, while showing low in vivo toxicity at a high dose. Based on these observations, it can be concluded that compound 1 is a promising anti-PDK1-3 lead that merits further investigation.


Subject(s)
Protein Serine-Threonine Kinases , Pyruvate Dehydrogenase Complex , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Oxidative Phosphorylation , Cell Division
SELECTION OF CITATIONS
SEARCH DETAIL