Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
1.
Nucleic Acids Res ; 52(11): 6629-6646, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38567728

ABSTRACT

Enzyme activity is determined by various different mechanisms, including posttranslational modifications and allosteric regulation. Allosteric activators are often metabolites but other molecules serve similar functions. So far, examples of long non-coding RNAs (lncRNAs) acting as allosteric activators of enzyme activity are missing. Here, we describe the function of mitolnc in cardiomyocytes, a nuclear encoded long non-coding RNA, located in mitochondria and directly interacting with the branched-chain ketoacid dehydrogenase (BCKDH) complex to increase its activity. The BCKDH complex is critical for branched-chain amino acid catabolism (BCAAs). Inactivation of mitolnc in mice reduces BCKDH complex activity, resulting in accumulation of BCAAs in the heart and cardiac hypertrophy via enhanced mTOR signaling. We found that mitolnc allosterically activates the BCKDH complex, independent of phosphorylation. Mitolnc-mediated regulation of the BCKDH complex constitutes an important additional layer to regulate the BCKDH complex in a tissue-specific manner, evading direct coupling of BCAA metabolism to ACLY-dependent lipogenesis.


Subject(s)
Amino Acids, Branched-Chain , Cardiomegaly , RNA, Long Noncoding , Animals , Allosteric Regulation , Mice , Cardiomegaly/metabolism , Cardiomegaly/genetics , Amino Acids, Branched-Chain/metabolism , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Myocytes, Cardiac/metabolism , Humans , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , Signal Transduction , Mice, Inbred C57BL , TOR Serine-Threonine Kinases/metabolism , Male , Mice, Knockout
2.
J Biol Chem ; 300(3): 105702, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301896

ABSTRACT

Elevated levels of branched chain amino acids (BCAAs) and branched-chain α-ketoacids are associated with cardiovascular and metabolic disease, but the molecular mechanisms underlying a putative causal relationship remain unclear. The branched-chain ketoacid dehydrogenase kinase (BCKDK) inhibitor BT2 (3,6-dichlorobenzo[b]thiophene-2-carboxylic acid) is often used in preclinical models to increase BCAA oxidation and restore steady-state BCAA and branched-chain α-ketoacid levels. BT2 administration is protective in various rodent models of heart failure and metabolic disease, but confoundingly, targeted ablation of Bckdk in specific tissues does not reproduce the beneficial effects conferred by pharmacologic inhibition. Here, we demonstrate that BT2, a lipophilic weak acid, can act as a mitochondrial uncoupler. Measurements of oxygen consumption, mitochondrial membrane potential, and patch-clamp electrophysiology show that BT2 increases proton conductance across the mitochondrial inner membrane independently of its inhibitory effect on BCKDK. BT2 is roughly sixfold less potent than the prototypical uncoupler 2,4-dinitrophenol and phenocopies 2,4-dinitrophenol in lowering de novo lipogenesis and mitochondrial superoxide production. The data suggest that the therapeutic efficacy of BT2 may be attributable to the well-documented effects of mitochondrial uncoupling in alleviating cardiovascular and metabolic disease.


Subject(s)
Lipogenesis , Metabolic Diseases , Mitochondrial Membranes , Protein Kinase Inhibitors , Reactive Oxygen Species , Humans , 2,4-Dinitrophenol/pharmacology , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Lipogenesis/drug effects , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Animals , Mice , Rats , Cell Line , Mitochondrial Membranes/drug effects , Cells, Cultured
3.
J Bacteriol ; 206(7): e0003324, 2024 07 25.
Article in English | MEDLINE | ID: mdl-38899896

ABSTRACT

Listeria monocytogenes is a foodborne bacterial pathogen that causes listeriosis. Positive regulatory factor A (PrfA) is a pleiotropic master activator of virulence genes of L. monocytogenes that becomes active upon the entry of the bacterium into the cytosol of infected cells. L. monocytogenes can survive and multiply at low temperatures; this is accomplished through the maintenance of appropriate membrane fluidity via branched-chain fatty acid (BCFA) synthesis. Branched-chain α-keto acid dehydrogenase (BKD), which is composed of four polypeptides encoded by lpd, bkdA1, bkdA2, and bkdB, is known to play a vital role in BCFA biosynthesis. Here, we constructed BKD-deficient Listeria strains by in-frame deletion of lpd, bkdA1, bkdA2, and bkdB genes. To determine the role in in vivo and in vitro, mouse model challenges, plaque assay in murine L2 fibroblast, and intracellular replication in J744A.1 macrophage were conducted. BKD-deficient strains exhibited defects in BCFA composition, virulence, and PrfA-regulon function within the host cells. Transcriptomics analysis revealed that the transcript level of the PrfA-regulon was lower in ΔbkdA1 strain than those in the wild-type. This study demonstrates that L. monocytogenes strains lacking BKD complex components were defective in PrfA-regulon function, and full activation of wild-type prfA may not occur within host cells in the absence of BKD. Further study will investigate the consequences of BKD deletion on PrfA function through altering BCFA catabolism.IMPORTANCEListeria monocytogenes is the causative agent of listeriosis, a disease with a high mortality rate. In this study, we have shown that the deletion of BKD can impact the function of PrfA and the PrfA-regulon. The production of virulence proteins within host cells is necessary for L. monocytogenes to promote its intracellular survival and is likely dependent on membrane integrity. We thus report a link between L. monocytogenes membrane integrity and the function of PrfA. This knowledge will increase our understanding of L. monocytogenes pathogenesis, which may provide insight into the development of antimicrobial agents.


Subject(s)
Bacterial Proteins , Listeria monocytogenes , Listeriosis , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/enzymology , Listeria monocytogenes/metabolism , Mice , Animals , Virulence , Listeriosis/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fatty Acids/biosynthesis , Fatty Acids/metabolism , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , Gene Expression Regulation, Bacterial , Macrophages/microbiology , Female , Cell Line
4.
J Biol Chem ; 299(11): 105333, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37827290

ABSTRACT

Branched chain α-ketoacid dehydrogenase complex (BCKDC) is the rate-limiting enzyme in branched chain amino acid (BCAA) catabolism, a metabolic pathway with great importance for human health. BCKDC belongs to the mitochondrial α-ketoacid dehydrogenase complex family, which also includes pyruvate dehydrogenase complex and oxoglutarate dehydrogenase complex. Here, we revealed that BCKDC can be substantially inhibited by reactive nitrogen species (RNS) via a mechanism similar to what we recently discovered with pyruvate dehydrogenase complex and oxoglutarate dehydrogenase complex-RNS can cause inactivating covalent modifications of the lipoic arm on its E2 subunit. In addition, we showed that such reaction between RNS and the lipoic arm of the E2 subunit can further promote inhibition of the E3 subunits of α-ketoacid dehydrogenase complexes. We examined the impacts of this RNS-mediated BCKDC inhibition in muscle cells, an important site of BCAA metabolism, and demonstrated that the nitric oxide production induced by cytokine stimulation leads to a strong inhibition of BCKDC activity and BCAA oxidation in myotubes and myoblasts. More broadly, nitric oxide production reduced the level of functional lipoic arms across the multiple α-ketoacid dehydrogenases and led to intracellular accumulation of their substrates (α-ketoacids), decrease of their products (acyl-CoAs), and a lower cellular energy charge. In sum, this work revealed a new mechanism for BCKDC regulation, demonstrated that RNS can generally inhibit all α-ketoacid dehydrogenases, which has broad physiological implications across multiple cell types, and elucidated the mechanistic connection between RNS-driven inhibitory modifications on the E2 and E3 subunits of α-ketoacid dehydrogenases.


Subject(s)
3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide) , Muscle Cells , Nitric Oxide , Reactive Nitrogen Species , Humans , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Ketoglutarate Dehydrogenase Complex , Muscle Cells/metabolism , Pyruvate Dehydrogenase Complex , Reactive Nitrogen Species/metabolism
5.
J Biol Chem ; 299(3): 102959, 2023 03.
Article in English | MEDLINE | ID: mdl-36717078

ABSTRACT

The mammalian mitochondrial branched-chain ketoacid dehydrogenase (BCKD) complex is a multienzyme complex involved in the catabolism of branched-chain amino acids. BCKD is regulated by the BCKD kinase, or BCKDK, which binds to the E2 subunit of BCKD, phosphorylates its E1 subunit, and inhibits enzymatic activity. Inhibition of the BCKD complex results in increased levels of branched-chain amino acids and branched-chain ketoacids, and this buildup has been associated with heart failure, type 2 diabetes mellitus, and nonalcoholic fatty liver disease. To find BCKDK inhibitors for potential treatment of these diseases, we performed both NMR and virtual fragment screening and identified tetrazole-bearing fragments that bind BCKDK at multiple sites. Through structure-based virtual screening expanding from these fragments, the angiotensin receptor blocker class antihypertension drugs and angiotensin receptor blocker-like compounds were discovered to be potent BCKDK inhibitors, suggesting potential new avenues for heart failure treatment combining BCKDK inhibition and antihypertension.


Subject(s)
3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide) , Angiotensin Receptor Antagonists , Humans , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Angiotensin Receptor Antagonists/pharmacology , Multienzyme Complexes/metabolism , Heart Failure , Hypertension
6.
J Inherit Metab Dis ; 47(1): 41-49, 2024 Jan.
Article in English | MEDLINE | ID: mdl-36880392

ABSTRACT

Maple syrup urine disease (MSUD) is rare autosomal recessive metabolic disorder caused by the dysfunction of the mitochondrial branched-chain 2-ketoacid dehydrogenase (BCKD) enzyme complex leading to massive accumulation of branched-chain amino acids and 2-keto acids. MSUD management, based on a life-long strict protein restriction with nontoxic amino acids oral supplementation represents an unmet need as it is associated with a poor quality of life, and does not fully protect from acute life-threatening decompensations or long-term neuropsychiatric complications. Orthotopic liver transplantation is a beneficial therapeutic option, which shows that restoration of only a fraction of whole-body BCKD enzyme activity is therapeutic. MSUD is thus an ideal target for gene therapy. We and others have tested AAV gene therapy in mice for two of the three genes involved in MSUD, BCKDHA and DBT. In this study, we developed a similar approach for the third MSUD gene, BCKDHB. We performed the first characterization of a Bckdhb-/- mouse model, which recapitulates the severe human phenotype of MSUD with early-neonatal symptoms leading to death during the first week of life with massive accumulation of MSUD biomarkers. Based on our previous experience in Bckdha-/- mice, we designed a transgene carrying the human BCKDHB gene under the control of a ubiquitous EF1α promoter, encapsidated in an AAV8 capsid. Injection in neonatal Bckdhb-/- mice at 1014 vg/kg achieved long-term rescue of the severe MSUD phenotype of Bckdhb-/- mice. These data further validate the efficacy of gene therapy for MSUD opening perspectives towards clinical translation.


Subject(s)
Maple Syrup Urine Disease , Animals , Humans , Mice , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/chemistry , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Maple Syrup Urine Disease/genetics , Maple Syrup Urine Disease/therapy , Maple Syrup Urine Disease/diagnosis , Phenotype , Quality of Life
7.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062842

ABSTRACT

Muscle wasting can be caused by nutrition deficiency and inefficient metabolism of amino acids, including Branched Chain Amino Acids (BCAAs). Branched Chain Amino Acids are a major contributor to the metabolic needs of healthy muscle and account for over a tenth of lean muscle mass. Branched chain alpha-ketoacid dehydrogenase (BCKD) is the rate limiting enzyme of BCAA metabolism. Inhibition of BCKD is achieved through a reversible phosphorylation event by Branched Chain a-ketoacid dehydrogenase kinase (BCKDK). Our study set out to determine the importance of BCKDK in the maintenance of skeletal muscle. We used the Gene Expression Omnibus Database to understand the role of BCKDK in skeletal muscle pathogenesis, including aging, muscular disease, and interrupted muscle metabolism. We found BCKDK expression levels were consistently decreased in pathologic conditions. These results were most consistent when exploring muscular disease followed by aging. Based on our findings, we hypothesize that decreased BCKDK expression alters BCAA catabolism and impacts loss of normal muscle integrity and function. Further research could offer valuable insights into potential therapeutic strategies for addressing muscle-related disorders.


Subject(s)
Amino Acids, Branched-Chain , Muscle, Skeletal , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Humans , Amino Acids, Branched-Chain/metabolism , Animals , Aging/metabolism , Aging/genetics , Muscular Diseases/metabolism , Muscular Diseases/pathology , Muscular Diseases/genetics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , Protein Kinases
8.
Am J Med Genet A ; 191(5): 1360-1365, 2023 05.
Article in English | MEDLINE | ID: mdl-36706222

ABSTRACT

Maple syrup urine disease (MSUD) is an inborn error of metabolism caused by the insufficient catabolism of branched-chain amino acids. BCKDHA, BCKDHB, DBT, and DLD encode the subunits of the branched-chain α-ketoacid dehydrogenase complex, which is responsible for the catabolism of these amino acids. Biallelic pathogenic variants in BCKDHA, BCKDHB, or DBT are characteristic of MSUD. In addition, a patient with a PPM1K defect was previously reported. PPM1K dephosphorylates and activates the enzyme complex. We report a patient with MSUD with mild findings and elevated BCAA levels carrying a novel homozygous start-loss variant in PPM1K. Our study offers further evidence that PPM1K variants cause mild MSUD.


Subject(s)
Maple Syrup Urine Disease , Protein Phosphatase 2C , Humans , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/chemistry , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Homozygote , Maple Syrup Urine Disease/diagnosis , Maple Syrup Urine Disease/genetics , Mutation , Protein Phosphatase 2C/genetics
9.
Cell Mol Life Sci ; 78(23): 7451-7468, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34718827

ABSTRACT

In human metabolism, pyruvate dehydrogenase complex (PDC) is one of the most intricate and large multimeric protein systems representing a central hub for cellular homeostasis. The worldwide used antiepileptic drug valproic acid (VPA) may potentially induce teratogenicity or a mild to severe hepatic toxicity, where the underlying mechanisms are not completely understood. This work aims to clarify the mechanisms that intersect VPA-related iatrogenic effects to PDC-associated dihydrolipoamide dehydrogenase (DLD; E3) activity. DLD is also a key enzyme of α-ketoglutarate dehydrogenase, branched-chain α-keto acid dehydrogenase, α-ketoadipate dehydrogenase, and the glycine decarboxylase complexes. The molecular effects of VPA will be reviewed underlining the data that sustain a potential interaction with DLD. The drug-associated effects on lipoic acid-related complexes activity may induce alterations on the flux of metabolites through tricarboxylic acid cycle, branched-chain amino acid oxidation, glycine metabolism and other cellular acetyl-CoA-connected reactions. The biotransformation of VPA involves its complete ß-oxidation in mitochondria causing an imbalance on energy homeostasis. The drug consequences as histone deacetylase inhibitor and thus gene expression modulator have also been recognized. The mitochondrial localization of PDC is unequivocal, but its presence and function in the nucleus were also demonstrated, generating acetyl-CoA, crucial for histone acetylation. Bridging metabolism and epigenetics, this review gathers the evidence of VPA-induced interference with DLD or PDC functions, mainly in animal and cellular models, and highlights the uncharted in human. The consequences of this interaction may have significant impact either in mitochondrial or in nuclear acetyl-CoA-dependent processes.


Subject(s)
Dihydrolipoamide Dehydrogenase/metabolism , Histone Deacetylase Inhibitors/adverse effects , Iatrogenic Disease , Pyruvate Dehydrogenase Complex/metabolism , Valproic Acid/adverse effects , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Acetyl Coenzyme A/biosynthesis , Acetylation , Animals , Glycine Dehydrogenase (Decarboxylating)/metabolism , Humans , Ketoglutarate Dehydrogenase Complex/metabolism , Ketone Oxidoreductases/metabolism , Liver/pathology , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Teratogens/metabolism
10.
Biochem J ; 478(4): 765-776, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33626142

ABSTRACT

Oxidation of branched-chain amino acids (BCAAs) is tightly regulated in mammals. We review here the distribution and regulation of whole-body BCAA oxidation. Phosphorylation and dephosphorylation of the rate-limiting enzyme, branched-chain α-ketoacid dehydrogenase complex directly regulates BCAA oxidation, and various other indirect mechanisms of regulation also exist. Most tissues throughout the body are capable of BCAA oxidation, and the flux of oxidative BCAA disposal in each tissue is influenced by three key factors: 1. tissue-specific preference for BCAA oxidation relative to other fuels, 2. the overall oxidative activity of mitochondria within a tissue, and 3. total tissue mass. Perturbations in BCAA oxidation have been implicated in many disease contexts, underscoring the importance of BCAA homeostasis in overall health.


Subject(s)
3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Animals , Bacterial Proteins/metabolism , Decarboxylation , Female , Forecasting , Heart Failure/metabolism , Humans , Insulin Resistance/physiology , Male , Maple Syrup Urine Disease/genetics , Maple Syrup Urine Disease/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/enzymology , Multienzyme Complexes , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Organ Specificity , Oxidation-Reduction , Phosphorylation , Plant Proteins/metabolism , Protein Processing, Post-Translational
11.
Int J Mol Sci ; 23(7)2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35409380

ABSTRACT

Branched chain amino acids (BCAAs), leucine, isoleucine and valine, are essential amino acids widely studied for their crucial role in the regulation of protein synthesis mainly through the activation of the mTOR signaling pathway and their emerging recognition as players in the regulation of various physiological and metabolic processes, such as glucose homeostasis. BCAA supplementation is primarily used as a beneficial nutritional intervention in chronic liver and kidney disease as well as in muscle wasting disorders. However, downregulated/upregulated plasma BCAAs and their defective catabolism in various tissues, mainly due to altered enzymatic activity of the first two enzymes in their catabolic pathway, BCAA aminotransferase (BCAT) and branched-chain α-keto acid dehydrogenase (BCKD), have been investigated in many nutritional and disease states. The current review focused on the underlying mechanisms of altered BCAA catabolism and its contribution to the pathogenesis of a numerous pathological conditions such as diabetes, heart failure and cancer. In addition, we summarize findings that indicate that the recovery of the dysregulated BCAA catabolism may be associated with an improved outcome and the prevention of serious disease complications.


Subject(s)
Amino Acids, Branched-Chain , Transaminases , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Humans , Leucine , Transaminases/metabolism
12.
J Biol Chem ; 295(46): 15597-15621, 2020 11 13.
Article in English | MEDLINE | ID: mdl-32878988

ABSTRACT

Branched-chain α-keto acids (BCKAs) are catabolites of branched-chain amino acids (BCAAs). Intracellular BCKAs are cleared by branched-chain ketoacid dehydrogenase (BCKDH), which is sensitive to inhibitory phosphorylation by BCKD kinase (BCKDK). Accumulation of BCKAs is an indicator of defective BCAA catabolism and has been correlated with glucose intolerance and cardiac dysfunction. However, it is unclear whether BCKAs directly alter insulin signaling and function in the skeletal and cardiac muscle cell. Furthermore, the role of excess fatty acids (FAs) in perturbing BCAA catabolism and BCKA availability merits investigation. By using immunoblotting and ultra-performance liquid chromatography MS/MS to analyze the hearts of fasted mice, we observed decreased BCAA-catabolizing enzyme expression and increased circulating BCKAs, but not BCAAs. In mice subjected to diet-induced obesity (DIO), we observed similar increases in circulating BCKAs with concomitant changes in BCAA-catabolizing enzyme expression only in the skeletal muscle. Effects of DIO were recapitulated by simulating lipotoxicity in skeletal muscle cells treated with saturated FA, palmitate. Exposure of muscle cells to high concentrations of BCKAs resulted in inhibition of insulin-induced AKT phosphorylation, decreased glucose uptake, and mitochondrial oxygen consumption. Altering intracellular clearance of BCKAs by genetic modulation of BCKDK and BCKDHA expression showed similar effects on AKT phosphorylation. BCKAs increased protein translation and mTORC1 activation. Pretreating cells with mTORC1 inhibitor rapamycin restored BCKA's effect on insulin-induced AKT phosphorylation. This study provides evidence for FA-mediated regulation of BCAA-catabolizing enzymes and BCKA content and highlights the biological role of BCKAs in regulating muscle insulin signaling and function.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Insulin/metabolism , Muscle, Skeletal/metabolism , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/antagonists & inhibitors , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/blood , Animals , Cell Line , Diet, High-Fat , Down-Regulation/drug effects , Insulin/pharmacology , Keto Acids/blood , Keto Acids/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Myocardium/metabolism , Palmitates/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
13.
Pharmacol Res ; 167: 105518, 2021 05.
Article in English | MEDLINE | ID: mdl-33636353

ABSTRACT

Catabolism of branched-chain amino acids (BCAAs) is affected by various physiological conditions and its abnormality is associated with glucose metabolism, heart disease, and neurological dysfunction. The first two steps of the BCAA metabolic pathway are common to the three BCAAs (leucine, isoleucine, and valine). The second step is an irreversible rate-limited reaction catalyzed by branched-chain α-keto acid dehydrogenase (BCKDH), which is bound to a specific kinase, BCKDH kinase (BDK), and inactivated by phosphorylation. Here, we investigated potential new BDK inhibitors and discovered valsartan, an angiotensin II type 1 receptor (AT1R) blocker, as a new BDK inhibitor. BCKDH phosphorylation and the BCKDH-BDK interaction were inhibited by valsartan in vitro. Valsartan administration in rats resulted in increased BCKDH activity by decreasing the dephosphorylated level of BCKDH complex, bound forms of BDK from BCKDH complex as well as decreased plasma BCAA concentrations. Valsartan is a novel BDK inhibitor that competes with ATP, via a different mechanism from allosteric inhibitors. The BDK inhibitor has been shown to preserve cardiac function in pressure overload-induced heart failure mice and to attenuate insulin resistance in obese mice. Our findings suggest that valsartan is a potent seed compound for developing a powerful BDK inhibitor and useful medication for treating heart failure and metabolic diseases with suppressed BCAA catabolism.


Subject(s)
Antihypertensive Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Valsartan/pharmacology , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Animals , Female , Protein Interaction Maps/drug effects , Rats , Rats, Sprague-Dawley
14.
Microb Cell Fact ; 20(1): 111, 2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34082758

ABSTRACT

BACKGROUND: Pamamycins are macrodiolides of polyketide origin which form a family of differently large homologues with molecular weights between 579 and 663. They offer promising biological activity against pathogenic fungi and gram-positive bacteria. Admittedly, production titers are very low, and pamamycins are typically formed as crude mixture of mainly smaller derivatives, leaving larger derivatives rather unexplored so far. Therefore, strategies that enable a more efficient production of pamamycins and provide increased fractions of the rare large derivatives are highly desired. Here we took a systems biology approach, integrating transcription profiling by RNA sequencing and intracellular metabolite analysis, to enhance pamamycin production in the heterologous host S. albus J1074/R2. RESULTS: Supplemented with L-valine, the recombinant producer S. albus J1074/R2 achieved a threefold increased pamamycin titer of 3.5 mg L-1 and elevated fractions of larger derivatives: Pam 649 was strongly increased, and Pam 663 was newly formed. These beneficial effects were driven by increased availability of intracellular CoA thioesters, the building blocks for the polyketide, resulting from L-valine catabolism. Unfavorably, L-valine impaired growth of the strain, repressed genes of mannitol uptake and glycolysis, and suppressed pamamycin formation, despite the biosynthetic gene cluster was transcriptionally activated, restricting production to the post L-valine phase. A deletion mutant of the transcriptional regulator bkdR, controlling a branched-chain amino acid dehydrogenase complex, revealed decoupled pamamycin biosynthesis. The regulator mutant accumulated the polyketide independent of the nutrient status. Supplemented with L-valine, the novel strain enabled the biosynthesis of pamamycin mixtures with up to 55% of the heavy derivatives Pam 635, Pam 649, and Pam 663: almost 20-fold more than the wild type. CONCLUSIONS: Our findings open the door to provide rare heavy pamamycins at markedly increased efficiency and facilitate studies to assess their specific biological activities and explore this important polyketide further.


Subject(s)
Macrolides/metabolism , Polyketides/metabolism , Streptomyces/genetics , Streptomyces/metabolism , Transcription Factors/genetics , Valine/metabolism , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Bacterial Proteins/genetics , Biosynthetic Pathways , Gene Expression Profiling/methods , Gene Expression Regulation, Bacterial , Genes, Bacterial , Industrial Microbiology , Metabolome , Multigene Family , Mutation
15.
Nutr Metab Cardiovasc Dis ; 31(11): 3210-3218, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34511290

ABSTRACT

BACKGROUND AND AIM: Circulating amino acids are modified by sex, body mass index (BMI) and insulin resistance (IR). However, whether the presence of genetic variants in branched-chain amino acid (BCAA) catabolic enzymes modifies circulating amino acids is still unknown. Thus, we determined the frequency of two genetic variants, one in the branched-chain aminotransferase 2 (BCAT2) gene (rs11548193), and one in the branched-chain ketoacid dehydrogenase (BCKDH) gene (rs45500792), and elucidated their impact on circulating amino acid levels together with clinical, anthropometric and biochemical parameters. METHODS AND RESULTS: We performed a cross-sectional comparative study in which we recruited 1612 young adults (749 women and 863 men) aged 19.7 ± 2.1 years and with a BMI of 24.9 ± 4.7 kg/m2. Participants underwent clinical evaluation and provided blood samples for DNA extraction and biochemical analysis. The single nucleotide polymorphisms (SNPs) were determined by allelic discrimination using real-time polymerase chain reaction (PCR). The frequencies of the less common alleles were 15.2 % for BCAT2 and 9.83 % for BCKDH. The subjects with either the BCAT2 or BCKDH SNPs displayed no differences in the evaluated parameters compared with subjects homozygotes for the most common allele at each SNP. However, subjects with both SNPs had higher body weight, BMI, blood pressure, glucose, and circulating levels of aspartate, isoleucine, methionine, and proline than the subjects homozygotes for the most common allele (P < 0.05, One-way ANOVA). CONCLUSION: Our findings suggest that the joint presence of both the BCAT2 rs11548193 and BCKDH rs45500792 SNPs induces metabolic alterations that are not observed in subjects without either SNP.


Subject(s)
3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , Amino Acids/blood , Minor Histocompatibility Antigens/genetics , Polymorphism, Single Nucleotide , Pregnancy Proteins/genetics , Transaminases/genetics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Adolescent , Age Factors , Biomarkers/blood , Blood Glucose/analysis , Blood Pressure , Body Mass Index , Cross-Sectional Studies , Female , Gene Frequency , Genetic Association Studies , Homozygote , Humans , Male , Mexico , Minor Histocompatibility Antigens/metabolism , Phenotype , Pregnancy Proteins/metabolism , Transaminases/metabolism , Young Adult
16.
Mol Genet Metab ; 129(3): 193-206, 2020 03.
Article in English | MEDLINE | ID: mdl-31980395

ABSTRACT

Over the past three decades, we studied 184 individuals with 174 different molecular variants of branched-chain α-ketoacid dehydrogenase activity, and here delineate essential clinical and biochemical aspects of the maple syrup urine disease (MSUD) phenotype. We collected data about treatment, survival, hospitalization, metabolic control, and liver transplantation from patients with classic (i.e., severe; n = 176), intermediate (n = 6) and intermittent (n = 2) forms of MSUD. A total of 13,589 amino acid profiles were used to analyze leucine tolerance, amino acid homeostasis, estimated cerebral amino acid uptake, quantitative responses to anabolic therapy, and metabolic control after liver transplantation. Standard instruments were used to measure neuropsychiatric outcomes. Despite advances in clinical care, classic MSUD remains a morbid and potentially fatal disorder. Stringent dietary therapy maintains metabolic variables within acceptable limits but is challenging to implement, fails to restore appropriate concentration relationships among circulating amino acids, and does not fully prevent cognitive and psychiatric disabilities. Liver transplantation eliminates the need for a prescription diet and safeguards patients from life-threatening metabolic crises, but is associated with predictable morbidities and does not reverse pre-existing neurological sequelae. There is a critical unmet need for safe and effective disease-modifying therapies for MSUD which can be implemented early in life. The biochemistry and physiology of MSUD and its response to liver transplantation afford key insights into the design of new therapies based on gene replacement or editing.


Subject(s)
3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , Amino Acids, Branched-Chain/metabolism , Biomarkers/blood , Leucine/blood , Liver Transplantation , Maple Syrup Urine Disease/diet therapy , Maple Syrup Urine Disease/therapy , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Adolescent , Adult , Child , Child, Preschool , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cohort Studies , Diet , Female , Homozygote , Humans , Infant , Leucine/metabolism , Male , Maple Syrup Urine Disease/genetics , Maple Syrup Urine Disease/metabolism , Mental Disorders/metabolism , Mental Disorders/physiopathology , Middle Aged , Phenotype
17.
Mol Cell Biochem ; 468(1-2): 169-183, 2020 May.
Article in English | MEDLINE | ID: mdl-32222880

ABSTRACT

Population data have consistently demonstrated a correlation between circulating branched-chain amino acids (BCAA) and insulin resistance. Most recently valine catabolite, 3-hydroxyisobutyrate, has emerged as a potential cause of BCAA-mediated insulin resistance; however, it is unclear if valine independently promotes insulin resistance. It is also unclear if excess valine influences the ability of cells to degrade BCAA. Therefore, this study investigated the effect of valine on muscle insulin signaling and related metabolism in vitro. C2C12 myotubes were treated with varying concentrations (0.5 mM-2 mM) of valine for up to 48 h. qRT-PCR and western blot were used to measure metabolic gene and protein expression, respectively. Insulin sensitivity (indicated by pAkt:Akt), metabolic gene and protein expression, and cell metabolism were also measured following valine treatment both with and without varying levels of insulin resistance. Mitochondrial and glycolytic metabolism were measured via oxygen consumption and extracellular acidification rate, respectively. Valine did not alter regulators of mitochondrial biogenesis or glycolysis; however, valine reduced branched-chain alpha-keto acid dehydrogenase a (Bckdha) mRNA (but not protein) expression which was exacerbated by insulin resistance. Valine treatment had no effect on pAkt:Akt following either acute or 48-h treatment, regardless of insulin stimulation or varying levels of insulin resistance. In conclusion, despite consistent population data demonstrating a relationship between circulating BCAA (and related metabolites) and insulin resistance, valine does not appear to independently alter insulin sensitivity or worsen insulin resistance in the myotube model of skeletal muscle.


Subject(s)
Amino Acids, Branched-Chain/drug effects , Insulin Resistance , Insulin/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Valine/pharmacology , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Animals , Cell Line , Cell Survival/drug effects , Glycolysis/drug effects , Insulin/pharmacology , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
18.
J Dairy Sci ; 103(3): 2847-2863, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31928756

ABSTRACT

Branched-chain amino acids (BCAA) are major components of milk protein and important precursors for nonessential AA. Thus, the BCAA transport and break-down play a key role in the metabolic adaptation to the high nutrient demands in lactation. However, in monogastrics, increased BCAA levels have been linked with obesity and certain metabolic disorders such as impaired insulin sensitivity. Our objective was to study the effect of over-conditioning at calving on plasma BCAA levels as well as the tissue abundance of the most relevant BCAA transporters and degrading enzymes in dairy cows during late pregnancy and early lactation. Thirty-eight Holstein cows were allocated 15 wk antepartum to either a normal- (NBCS) or over-conditioned (HBCS) group, receiving 6.8 or 7.2 MJ of NEL/kg of DM, respectively, during late lactation to reach the targeted differences in body condition score (BCS) and back fat thickness (BFT; NBCS: BCS <3.5, BFT <1.2 cm; HBCS: BCS >3.75, BFT >1.4 cm) until dry-off. During the dry period and next lactation, cows were fed the same diets, whereby differences in BCS and BFT were maintained: prepartum means were 3.16 ± 0.06 and 1.03 ± 0.07 cm (NBCS) vs. 3.77 ± 0.08 and 1.89 ± 0.11 cm (HBCS), postpartum means were 2.89 ± 0.06 and 0.81 ± 0.05 cm (NBCS) vs. 3.30 ± 0.06 and 1.38 ± 0.08 cm (HBCS). Blood and biopsies from liver, semitendinosus muscle, and subcutaneous adipose tissue (scAT) were sampled at d 49 antepartum, 3, 21, and 84 postpartum. Free BCAA were analyzed and the mRNA abundance of solute carrier family 1 member 5 (SLC1A5), SLC7A5, and SLC38A2 as well as branched-chain aminotransferase 2 (BCAT2), branched-chain α-keto acid dehydrogenase E1α (BCKDHA), and branched-chain α-keto acid dehydrogenase E1ß (BCKDHB) as well as the protein abundance of BCKDHA were assessed. Concentrations of all BCAA changed with time, most markedly in HBCS cows, with a nadir around calving. Apart from Ile, neither individual nor total BCAA differed between groups. The HBCS group had greater BCKDHA mRNA as well as higher prepartum BCKDHA protein abundance in scAT than NBCS cows, pointing to a greater oxidative capacity for the irreversible degradation of BCAA transamination products in scAT of over-conditioned cows. Prepartum hepatic BCKDHA protein abundance was lower in HBCS than in NBCS cows. In both groups, SLC1A5, SLC7A5, and BCAT2 mRNA were most abundant in scAT, whereas SLC38A2 was higher in scAT and muscle compared with liver, and BCKDHA and BCKDHB mRNA were greatest in liver and muscle, respectively. Our results indicate that scAT may be a major site of BCAA uptake and initial catabolism, with the former, however, being independent of BCS and time relative to calving in dairy cows.


Subject(s)
3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Cattle/physiology , Milk/chemistry , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Amino Acids, Branched-Chain/blood , Animals , Cattle/genetics , Diet/veterinary , Female , Lactation , Liver/metabolism , Muscle, Skeletal/enzymology , Postpartum Period , Pregnancy , RNA, Messenger/genetics , Subcutaneous Fat/enzymology
19.
Am J Physiol Endocrinol Metab ; 316(1): E73-E85, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30422704

ABSTRACT

Increased circulating branched-chain amino acids (BCAAs) have been involved in the pathogenesis of obesity and insulin resistance (IR). However, evidence relating berberine (BBR), gut microbiota, BCAAs, and IR is limited. Here, we showed that BBR could effectively rectify steatohepatitis and glucose intolerance in high-fat diet (HFD)-fed mice. BBR reorganized gut microbiota populations under both the normal chow diet (NCD) and HFD. Particularly, BBR noticeably decreased the relative abundance of BCAA-producing bacteria, including order Clostridiales; families Streptococcaceae, Clostridiaceae, and Prevotellaceae; and genera Streptococcus and Prevotella. Compared with the HFD group, predictive metagenomics indicated a reduction in the proportion of gut microbiota genes involved in BCAA biosynthesis but the enrichment genes for BCAA degradation and transport by BBR treatment. Accordingly, the elevated serum BCAAs of HFD group were significantly decreased by BBR. Furthermore, the Western blotting results implied that BBR could promote the BCAA catabolism in the liver and epididymal white adipose tissues of HFD-fed mice by activation of the multienzyme branched-chain α-ketoacid dehydrogenase complex (BCKDC), whereas by inhibition of the phosphorylation state of BCKDHA (E1α subunit) and branched-chain α-ketoacid dehydrogenase kinase (BCKDK). The ex vivo assay further confirmed that BBR could increase BCAA catabolism in both AML12 hepatocytes and 3T3-L1 adipocytes. Finally, data from healthy subjects and diabetics confirmed that BBR could improve glycemic control and modulate circulating BCAAs. Together, our findings clarified BBR improving IR associated not only with gut microbiota alteration in BCAA biosynthesis but also with BCAA catabolism in liver and adipose tissues.


Subject(s)
Adipose Tissue, White/drug effects , Amino Acids, Branched-Chain/drug effects , Berberine/pharmacology , Dysbiosis/metabolism , Gastrointestinal Microbiome/drug effects , Insulin Resistance , Liver/drug effects , Obesity/metabolism , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Adult , Amino Acids, Branched-Chain/metabolism , Animals , Diabetes Mellitus/metabolism , Diet, High-Fat , Fatty Liver , Female , Gastrointestinal Microbiome/genetics , Glucose Intolerance , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/metabolism , Male , Metagenomics , Mice , Middle Aged , Protein Kinases
20.
Clin Exp Immunol ; 195(1): 25-34, 2019 01.
Article in English | MEDLINE | ID: mdl-30099750

ABSTRACT

Primary biliary cholangitis (PBC) is a multi-factorial disease caused by the interaction of both genetic predisposition and environmental triggers. Bacterial infection has been investigated most intensively, both epidemiologically and experimentally, as a prime environmental aetiology in PBC. The association of recurrent history of urinary tract infection (UTI) with PBC has been frequently confirmed by several large-scale, case-control studies, despite variation in geographic area or case-finding methods. Escherichia coli is a predominant pathogen in most cases with UTI. Animal studies and molecular mimicry analysis between the human and E. coli E2 subunit of the 2-oxo-acid dehydrogenase complexes demonstrated that E. coli infection is a key factor in breaking immunological tolerance against the mitochondria, resulting in the production of anti-mitochondrial autoantibodies (AMA), the disease-specific autoantibodies of PBC. Novosphingobium aromaticivorans, a ubiquitous xenobiotic-metabolizing bacterium, is another candidate which may be involved in the aetiology of PBC. Meanwhile, improved environmental hygiene and increased prevalence of PBC, especially in males, may argue against the aetiological role of bacterial infection in PBC. Multiple mechanisms can result in the loss of tolerance to mitochondrial autoantigens in PBC; nonetheless, bacterial infection is probably one of the dominant pathways, especially in female patients. Notably, there is a rising prevalence of male patients with PBC. With increasing exposure to environmental xenobiotics in both genders, studies directed towards identifying the environmental culprit with systematically designed case-control studies are much needed to further determine the environmental factors and role of bacterial infections in PBC.


Subject(s)
Alphaproteobacteria/physiology , Autoimmunity , Bacterial Infections/microbiology , Liver Cirrhosis, Biliary/microbiology , Urinary Tract Infections/microbiology , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/immunology , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Animals , Autoantibodies/metabolism , Bacterial Infections/immunology , Female , Gene-Environment Interaction , Humans , Immune Tolerance , Liver Cirrhosis, Biliary/immunology , Male , Mitochondria/immunology , Molecular Mimicry , Urinary Tract Infections/immunology
SELECTION OF CITATIONS
SEARCH DETAIL