Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
J Inherit Metab Dis ; 46(3): 482-519, 2023 05.
Article in English | MEDLINE | ID: mdl-36221165

ABSTRACT

Glutaric aciduria type 1 is a rare inherited neurometabolic disorder of lysine metabolism caused by pathogenic gene variations in GCDH (cytogenic location: 19p13.13), resulting in deficiency of mitochondrial glutaryl-CoA dehydrogenase (GCDH) and, consequently, accumulation of glutaric acid, 3-hydroxyglutaric acid, glutaconic acid and glutarylcarnitine detectable by gas chromatography/mass spectrometry (organic acids) and tandem mass spectrometry (acylcarnitines). Depending on residual GCDH activity, biochemical high and low excreting phenotypes have been defined. Most untreated individuals present with acute onset of striatal damage before age 3 (to 6) years, precipitated by infectious diseases, fever or surgery, resulting in irreversible, mostly dystonic movement disorder with limited life expectancy. In some patients, striatal damage develops insidiously. In recent years, the clinical phenotype has been extended by the finding of extrastriatal abnormalities and cognitive dysfunction, preferably in the high excreter group, as well as chronic kidney failure. Newborn screening is the prerequisite for pre-symptomatic start of metabolic treatment with low lysine diet, carnitine supplementation and intensified emergency treatment during catabolic episodes, which, in combination, have substantially improved neurologic outcome. In contrast, start of treatment after onset of symptoms cannot reverse existing motor dysfunction caused by striatal damage. Dietary treatment can be relaxed after the vulnerable period for striatal damage, that is, age 6 years. However, impact of dietary relaxation on long-term outcomes is still unclear. This third revision of evidence-based recommendations aims to re-evaluate previous recommendations (Boy et al., J Inherit Metab Dis, 2017;40(1):75-101; Kolker et al., J Inherit Metab Dis 2011;34(3):677-694; Kolker et al., J Inherit Metab Dis, 2007;30(1):5-22) and to implement new research findings on the evolving phenotypic diversity as well as the impact of non-interventional variables and treatment quality on clinical outcomes.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Brain Diseases, Metabolic , Humans , Glutaryl-CoA Dehydrogenase , Lysine/metabolism , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/therapy , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/therapy , Glutarates/metabolism
2.
Neuroscience ; 471: 115-132, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34333063

ABSTRACT

D-2-hydroxyglutaric acid (D-2-HG) accumulates and is the biochemical hallmark of D-2-hydroxyglutaric acidurias (D-2-HGA) types I and II, which comprehend two inherited neurometabolic diseases with severe cerebral abnormalities. Since the pathogenesis of these diseases is poorly established, we tested whether D-2-HG could be neurotoxic to neonatal rats. D-2-HG intracerebroventricular administration caused marked vacuolation in cerebral cortex and striatum. In addition, glial fibrillary acidic protein (GFAP), S-100 calcium binding protein B (S100B) and ionized calcium-binding adapter molecule 1 (Iba-1) staining was increased in both brain structures, suggesting glial reactivity and microglial activation. D-2-HG also provoked a reduction of NeuN-positive cells in cerebral cortex, signaling neuronal death. Considering that disturbances in redox homeostasis and energy metabolism may be involved in neuronal damage and glial reactivity, we assessed whether D-2-HG could induce oxidative stress and bioenergetics impairment. D-2-HG treatment significantly augmented reactive oxygen and nitrogen species generation, provoked lipid peroxidation and protein oxidative damage, diminished glutathione concentrations and augmented superoxide dismutase and catalase activities in cerebral cortex. Increased reactive oxygen species generation, lipoperoxidation and protein oxidation were also found in striatum. Furthermore, the antagonist of NMDA glutamate receptor MK-801 and the antioxidant melatonin were able to prevent most of D-2-HG-induced pro-oxidant effects, implying the participation of these receptors in D-2-HG-elicited oxidative damage. Our results also demonstrated that D-2-HG markedly reduced the respiratory chain complex IV and creatine kinase activities. It is presumed that these deleterious pathomechanisms caused by D-2-HGA may be involved in the brain abnormalities characteristic of early-infantile onset D-2-HGA.


Subject(s)
Microglia , Oxidative Stress , Animals , Animals, Newborn , Cerebral Cortex , Energy Metabolism , Glutarates , Rats
3.
Int J Biol Macromol ; 166: 851-860, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33161076

ABSTRACT

We report a facile approach for the preparation of protein conjugated glutaric acid functionalized Fe3O4 magnetic nanoparticles (Pro-Glu-MNPs), having improved colloidal stability and heating efficacy. The Pro-Glu-MNPs were prepared by covalent conjugation of BSA protein onto the surface of glutaric acid functionalized Fe3O4 magnetic nanoparticles (Glu-MNPs) obtained through thermal decomposition. XRD and TEM analyses confirmed the formation of crystalline Fe3O4 nanoparticles of average size ~5 nm, whereas the conjugation of BSA protein to them was evident from XPS, FTIR, TGA, DLS and zeta-potential measurements. These Pro-Glu-MNPs showed good colloidal stability in different media (water, phosphate buffer saline, cell culture medium) and exhibited room temperature superparamagnetism with good magnetic field responsivity towards the external magnet. The induction heating studies revealed that the heating efficacy of these Pro-Glu-MNPs was strongly reliant on the particle concentration and their stabilizing media. In addition, they showed enhanced heating efficacy over Glu-MNPs as surface passivation by protein offers colloidal stability to them as well as prevents their aggregation under AC magnetic field. Further, Pro-Glu-MNPs are biocompatible towards normal cells and showed substantial cellular internalization in cancerous cells, suggesting their potential application in hyperthermia therapy.


Subject(s)
Hyperthermia, Induced/methods , Magnetic Iron Oxide Nanoparticles/chemistry , Nanoconjugates/chemistry , Serum Albumin, Bovine/chemistry , Glutarates/chemistry , HeLa Cells , Humans , MCF-7 Cells , Protein Stability
4.
Biochem Biophys Res Commun ; 533(4): 1393-1399, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33092792

ABSTRACT

Cytosolic carboxypeptidases (CCPs) comprise a unique subfamily of M14 carboxypeptidases and are erasers of the reversible protein posttranslational modification- polyglutamylation. Potent inhibitors for CCPs may serve as leading compounds targeting imbalanced polyglutamylation. However, no efficient CCP inhibitor has yet been reported. Here, we showed that 2-phosphonomethylpentanedioic acid (2-PMPA), a potent inhibitor of the distant M28 family member glutamate carboxypeptidase II (GCPII), rather than the typical M14 inhibitor 2-benzylsuccinic acid, could efficiently inhibit CCP activities. 2-PMPA inhibited the recombinant Nna1 (a.k.a. CCP1) for hydrolyzing a synthetic peptide in a mixed manner, with Ki and Ki' being 0.11 µM and 0.24 µM respectively. It inhibited Nna1 for deglutamylating tubulin, the best-known polyglutamylated protein, with an IC50 of 0.21 mM. Homology modeling predicted that the R-form of 2-PMPA is more favorable to bind Nna1, unlike that GCPII prefers to S-form. This work for the first time identified a potent inhibitor for CCP family.


Subject(s)
Glutamate Carboxypeptidase II/antagonists & inhibitors , Organophosphorus Compounds/pharmacology , Protease Inhibitors/pharmacology , Carboxypeptidases/antagonists & inhibitors , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Cytosol/enzymology , Drug Evaluation, Preclinical/methods , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Glutamate Carboxypeptidase II/chemistry , Glutamate Carboxypeptidase II/metabolism , Glutarates/pharmacology , Kinetics , Molecular Docking Simulation , Organophosphorus Compounds/chemistry , Protease Inhibitors/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine-Type D-Ala-D-Ala Carboxypeptidase/genetics , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism , Succinic Acid/pharmacology
5.
J Nutr ; 150(Suppl 1): 2556S-2560S, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33000154

ABSTRACT

Lysine is an essential amino acid, and inherited diseases of its metabolism therefore represent defects of lysine catabolism. Although some of these enzyme defects are not well described yet, glutaric aciduria type I (GA1) and antiquitin (2-aminoadipic-6-semialdehyde dehydrogenase) deficiency represent the most well-characterized diseases. GA1 is an autosomal recessive disorder due to a deficiency of glutaryl-CoA dehydrogenase. Untreated patients exhibit early onset macrocephaly and may present a neurological deterioration with regression and movement disorder at the time of a presumably "benign" infection most often during the first year of life. This is associated with a characteristic neuroimaging pattern with frontotemporal atrophy and striatal injuries. Diagnosis relies on the identification of glutaric and 3-hydroxyglutaric acid in urine along with plasma glutarylcarnitine. Treatment consists of a low-lysine diet aiming at reducing the putatively neurotoxic glutaric and 3-hydroxyglutaric acids. Additional therapeutic measures include administration of l-carnitine associated with emergency measures at the time of intercurrent illnesses aiming at preventing brain injury. Early treated (ideally through newborn screening) patients exhibit a favorable long-term neurocognitive outcome, whereas late-treated or untreated patients may present severe neurocognitive irreversible disabilities. Antiquitin deficiency is the most common form of pyridoxine-dependent epilepsy. α-Aminoadipic acid semialdehyde (AASA) and Δ-1-piperideine-6-carboxylate (P6C) accumulate proximal to the enzymatic block. P6C forms a complex with pyridoxal phosphate (PLP), a key vitamer of pyridoxine, thereby reducing PLP bioavailability and subsequently causing epilepsy. Urinary AASA is a biomarker of antiquitin deficiency. Despite seizure control, only 25% of the pyridoxine-treated patients show normal neurodevelopment. Low-lysine diet and arginine supplementation are proposed in some patients with decrease of AASA, but the impact on neurodevelopment is unclear. In summary, GA1 and antiquitin deficiency are the 2 main human defects of lysine catabolism. Both include neurological impairment. Lysine dietary restriction is a key therapy for GA1, whereas its benefits in antiquitin deficiency appear less clear.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic, Inborn/metabolism , Brain Diseases, Metabolic/metabolism , Brain/metabolism , Epilepsy/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Lysine/metabolism , 2-Aminoadipic Acid/analogs & derivatives , 2-Aminoadipic Acid/metabolism , Aldehyde Dehydrogenase/metabolism , Amino Acid Metabolism, Inborn Errors/therapy , Arginine/therapeutic use , Brain/pathology , Brain Diseases, Metabolic/therapy , Brain Diseases, Metabolic, Inborn/therapy , Carnitine/analogs & derivatives , Carnitine/metabolism , Carnitine/therapeutic use , Epilepsy/therapy , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/therapy , Pyridoxal Phosphate/metabolism , Pyridoxine/metabolism , Pyridoxine/therapeutic use
6.
Brain Res ; 1749: 147145, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33035499

ABSTRACT

Degeneration of striatal neurons and cortical atrophy are pathological characteristics of glutaric acidemia type I (GA-I), a disease characterized by accumulation of glutaric acid (GA). The mechanisms that lead to neuronal loss and cognitive impairment are still unclear. The purpose of this study was to verify if acute exposure to GA during the neonatal period is sufficient to trigger apoptotic processes and lead to learning delay in early and late period. Besides, whether N-acetylcysteine (NAC) would protect against impairment induced by GA. Pups mice received a dose of GA (2.5 µmol/ g) or saline, 12 hs after birth, and were treated with NAC (250 mg/kg) or saline, up to 21th day of life. Although GA exhibited deficits in the procedural and working memories in 21 and 40-day-old mice, NAC protected against cognitive impairment. In striatum and cortex, NAC prevented glial cells activation (GFAP and Iba-1), decreased NGF, Bcl-2 and NeuN, the increase of lipid peroxidation and PARP induced by GA in both ages. NAC protected against increased p75NTR induced by GA, but not in cortex of 21-day-old mice. Thus, we showed that the integrity of striatal and cortical pathways has an important role for learning and suggested that sustained glial reactivity in neonatal period can be an initial trigger for delay of cognitive development. Furthermore, NAC protected against cognitive impairment induced by GA. This work shows that early identification of the alterations induced by GA is important to avoid future clinical complications and suggest that NAC could be an adjuvant treatment for this acidemia.


Subject(s)
Acetylcysteine/pharmacology , Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Glutarates/pharmacology , Maze Learning/drug effects , Receptors, Nerve Growth Factor/metabolism , Animals , Apoptosis/drug effects , Cerebral Cortex/metabolism , Cognition/drug effects , Corpus Striatum/metabolism , Lipid Peroxidation/drug effects , Male , Mice , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects
7.
Toxicol In Vitro ; 65: 104753, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31857147

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive neoplasm correlated with human herpesvirus 8 (HHV8). Metabolic reprogramming is a hallmark of cancers. The alterations in cellular metabolism are important to the survival of HHV8 latently infected cells. Pyruvate dehydrogenase (PDH) controls the flux of metabolites between glycolysis and the tricarboxylic acid cycle (TCA cycle) and is a key enzyme in cancer metabolic reprogramming. Glutaminolysis is required for the survival of PEL cells. Glutamate dehydrogenase 1 (GDH1) converts glutamate into α-ketoglutarate supplying the TCA cycle with intermediates to support anaplerosis. Previously we have observed that epigallocatechin-3-gallate (EGCG) can induce PEL cell death and N-acetyl cysteine (NAC) attenuates EGCG induced PEL cell death. In this study, results showed that EGCG upregulated the expression of glucose transporter GLUT3, and reduced the expression of pyruvate dehydrogenase E1-alpha (PDHA1), the major regulator of PDH, and GDH1. NAC could partially reverse the effects of EGCG in PEL cells. Overexpression of PDHA1 in PEL cells or supplement of α-ketoglutarate attenuated EGCG induced cell death. EGCG also reduced the levels of oncometabolite D-2-hydroxyglutarate (D2HG). These results suggest that EGCG may modulate the metabolism of PEL cells leading to cell death.


Subject(s)
Catechin/analogs & derivatives , Herpesvirus 8, Human , Lymphoma, Primary Effusion/metabolism , Metabolic Networks and Pathways/drug effects , Pyruvate Dehydrogenase (Lipoamide)/genetics , Catechin/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/drug effects , Glutarates/metabolism , Humans , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology
8.
BMC Pediatr ; 19(1): 344, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31597564

ABSTRACT

BACKGROUND: Mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (mHS) deficiency is an autosomal recessive inborn error of metabolism, which will give rise to failure of ketogenesis in liver during illness or fasting. It is a very rare disease with only a few patients reported worldwide, most of which had a good prognosis after proper therapies. CASE PRESENTATION: We report a 9-month-old boy with mHS deficiency presenting with unusually severe and persistent acidosis after diarrhea and reduced oral food intake. The metabolic acidosis persisted even after supplementation with sugar and alkaline solution. Blood purification and assisted respiration alleviated symptoms, but a second onset induced by respiratory infection several days later led to multiple organ failure and death. Urine organic acid analysis during the acute episode revealed a complex pattern of ketogenic dicarboxylic and 3-hydroxydicarboxylic aciduria with prominent elevation of glutaric acid and adipic acid, which seem to be specific to mHS deficiency. Plasma acylcarnitine analysis revealed elevated 3-hydroxybutyrylcarnitine and acetylcarnitine. This is the first report of elevated 3-hydroxybutyrylcarnitine in mHS deficiency. Whole exome sequencing revealed a novel compound heterozygous mutation in HMGCS2 (c.100C > T and c.1465delA). CONCLUSION: This severe case suggests the need for patients with mHS deficiency to avoid recurrent illness because it can induce severe metabolic crisis, possibly leading to death. Such patients may also require special treatment, such as blood purification. Urine organic acid profile during the acute episode may give a hint to the disease.


Subject(s)
Acidosis/genetics , Acyl Coenzyme A/deficiency , Hydroxymethylglutaryl-CoA Synthase/genetics , Mitochondria/enzymology , Mutation/genetics , Acidosis/therapy , Acidosis/urine , Adipates/urine , Carnitine/analogs & derivatives , Carnitine/blood , Carnitine/urine , Diarrhea/complications , Dicarboxylic Acids/urine , Fatal Outcome , Frameshift Mutation/genetics , Glutarates/urine , Humans , Infant , Male , Multiple Organ Failure/complications , Respiratory Tract Infections/complications , Exome Sequencing
9.
Mater Sci Eng C Mater Biol Appl ; 98: 65-73, 2019 May.
Article in English | MEDLINE | ID: mdl-30813069

ABSTRACT

Since Fe3O4 nanoparticles synthesized by plant extracts possess good bio-compatibility and superparamagnetic properties, the possibility of these could be used as a carrier in drug delivery. In this work, doxorubicin hydrochloride (DOX), an anti-cancer drug, loaded on glutaric anhydride-functionalized magnetic nanoparticles (Fe3O4@SiO2-Glu) was investigated at varying pH values for effective drug delivery. Various factors affecting the adsorption of DOX onto the Fe3O4@SiO2-Glu were examined, where the adsorption efficiency of DOX reached 92% at a concentration of 20 mg/L employing 10 mg of Fe3O4@SiO2-Glu at 303 K in pH 7.4. However, the adsorption efficiency of DOX was decreased to 18% at acidic pH value down to 3.0, implicating that the drug releasing process was controlled by pH. Adsorption kinetics was fitting to pseudo-second-order and the isothermal adsorption conformed to Freundlich isotherm. The morphology and surface composition of the synthesized Fe3O4@SiO2-Glu were characterized by SEM, TEM, and N2 adsorption/desorption isotherms, revealing that the specific surface area being 62.6 m2/g and the size ranging from ~30 to 50 nm. The zeta potential results indicated that Fe3O4@SiO2-Glu were negatively charged in various pH from 3 to 8.5. Characterizations by FTIR and UV-Vis techniques suggested that the DOX was absorbed and it can be delivered by Fe3O4@SiO2-Glu.


Subject(s)
Anhydrides/chemistry , Doxorubicin/chemistry , Drug Carriers/chemistry , Glutarates/chemistry , Magnetite Nanoparticles/chemistry , Silicon Dioxide/chemistry , Adsorption , Humans , Spectroscopy, Fourier Transform Infrared
10.
Biotechnol Bioeng ; 116(2): 333-341, 2019 02.
Article in English | MEDLINE | ID: mdl-30450795

ABSTRACT

Glutaric acid is a promising alternative chemical to phthalate plasticizer since it can be produced by the bioconversion of lysine. Though, recent studies have enabled the high-yield production of its precursor, 5-aminovaleric acid (AMV), glutaric acid production via the AMV pathway has been limited by the need for cofactors. Introduction of NAD(P)H oxidase (Nox) with GabTD enzyme remarkably diminished the demand for oxidized nicotinamide adenine dinucleotide (NAD+ ). Supply of oxygen through vigorous shaking had a significant effect on the conversion of AMV with a reduced requirement of NAD + . A high conversion rate was achieved in Nox coupled GabTD reaction under optimized expression vector, terrific broth (TB), and pH 8.5 at high cell density. Supplementary expression of GabD resulted in the production of 353 ± 35 mM glutaric acid with 88.3 ± 8.7% conversion from 400 mM AMV. Moreover, the reaction with a higher concentration of AMV could produce 528 ± 21 mM glutaric acid with 66.0 ± 2.7% conversion. In addition, the co-biotransformation strategy of GabTD and DavBA whole cells could produce 282 mM glutaric acid with 70.8% conversion from lysine, compared to the 111 mM glutaric acid yield from the combined GabTD-DavBA system.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Glutarates/metabolism , Lysine/metabolism , Metabolic Engineering/methods , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/metabolism , Succinate-Semialdehyde Dehydrogenase/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Biotransformation , Escherichia coli/genetics , Recombinant Proteins/metabolism
11.
Mol Neurobiol ; 56(7): 4945-4959, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30421167

ABSTRACT

Dysfunction of basal ganglia neurons is a characteristic of glutaric acidemia type I (GA-I), an autosomal recessive inherited neurometabolic disease characterized by deficiency of glutaryl-CoA dehydrogenase (GCDH) and accumulation of glutaric acid (GA). The affected patients present clinical manifestations such as motor dysfunction and memory impairment followed by extensive striatal neurodegeneration. Knowing that there is relevant striatal dysfunction in GA-I, the purpose of the present study was to verify the performance of young rats chronically injected with GA in working and procedural memory test, and whether N-acetylcysteine (NAC) would protect against impairment induced by GA. Rat pups were injected with GA (5 µmol g body weight-1, subcutaneously; twice per day; from the 5th to the 28th day of life) and were supplemented with NAC (150 mg/kg/day; intragastric gavage; for the same period). We found that GA injection caused delay procedural learning; increase of cytokine concentration, oxidative markers, and caspase levels; decrease of antioxidant defenses; and alteration of acetylcholinesterase (AChE) activity. Interestingly, we found an increase in glial cell immunoreactivity and decrease in the immunoreactivity of nuclear factor-erythroid 2-related factor 2 (Nrf2), nicotinic acetylcholine receptor subunit alpha 7 (α7nAChR), and neuronal nuclei (NeuN) in the striatum. Indeed, NAC administration improved the cognitive performance, ROS production, neuroinflammation, and caspase activation induced by GA. NAC did not prevent neuronal death, however protected against alterations induced by GA on Iba-1 and GFAP immunoreactivities and AChE activity. Then, this study suggests possible therapeutic strategies that could help in GA-I treatment and the importance of the striatum in the learning tasks.


Subject(s)
Acetylcysteine/therapeutic use , Cholinergic Neurons/drug effects , Glutarates/toxicity , Maze Learning/drug effects , Memory Disorders/prevention & control , Neuroglia/drug effects , Acetylcysteine/pharmacology , Animals , Cholinergic Neurons/metabolism , Male , Maze Learning/physiology , Memory Disorders/chemically induced , Memory Disorders/metabolism , Neuroglia/metabolism , Rats , Rats, Wistar
12.
Cell ; 175(1): 101-116.e25, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30220459

ABSTRACT

IDH1 mutations are common in low-grade gliomas and secondary glioblastomas and cause overproduction of (R)-2HG. (R)-2HG modulates the activity of many enzymes, including some that are linked to transformation and some that are probably bystanders. Although prior work on (R)-2HG targets focused on 2OG-dependent dioxygenases, we found that (R)-2HG potently inhibits the 2OG-dependent transaminases BCAT1 and BCAT2, likely as a bystander effect, thereby decreasing glutamate levels and increasing dependence on glutaminase for the biosynthesis of glutamate and one of its products, glutathione. Inhibiting glutaminase specifically sensitized IDH mutant glioma cells to oxidative stress in vitro and to radiation in vitro and in vivo. These findings highlight the complementary roles for BCATs and glutaminase in glutamate biosynthesis, explain the sensitivity of IDH mutant cells to glutaminase inhibitors, and suggest a strategy for maximizing the effectiveness of such inhibitors against IDH mutant gliomas.


Subject(s)
Glioma/metabolism , Glutamic Acid/biosynthesis , Transaminases/physiology , Cell Line, Tumor , Glioma/physiopathology , Glutamic Acid/drug effects , Glutarates/metabolism , Glutarates/pharmacology , Homeostasis/drug effects , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/physiology , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/physiology , Mutation , Oxidation-Reduction/drug effects , Pregnancy Proteins/genetics , Pregnancy Proteins/physiology , Transaminases/antagonists & inhibitors , Transaminases/genetics
13.
IEEE Trans Nanobioscience ; 17(4): 394-401, 2018 10.
Article in English | MEDLINE | ID: mdl-29994125

ABSTRACT

As one of the new posttranslational modification, lysine glutarylation has been identified in both prokaryotic and eukaryotic cells. These glutarylated proteins are involved in various cellular functions, such as translation, metabolism, and exhibited diverse subcellular localizations. Experimental identification of lysine glutarylation sites was founded in 2014 and also identified its deglutarylase sirturn 5(SIRT 5). Computational prediction of lysine glutarylation could be a complementary way to the experimental technique. In this work, the lysine glutarylation predictor iGlu-Lys has been developed based on the machine learning scheme. We have selected the best feature scheme which took the amino acid pair order and special-position information into account from four constructions. The machine learning algorithm support vector machine has been adopted and its performance has been measured for different window length of peptides. In the 10-fold cross-validation with window length 19, the AUC and MCC were 0.8944 and 0.5098, respectively. Different ROC curves in 6-, 8-, and 10-fold cross-validations were very close which illustrated the robustness of our predictor. The results of iGLu-Lys were better than the existing method GlutPred. Meanwhile, a free webserver for iGlu-Lys is accessible at http://app.aporc.org/iGlu-Lys/.


Subject(s)
Computational Biology/methods , Lysine , Protein Processing, Post-Translational/physiology , Algorithms , Databases, Protein , Glutarates , Lysine/chemistry , Lysine/metabolism , Proteins/chemistry , Proteins/metabolism , ROC Curve , Sequence Analysis, Protein , Software
14.
Sci Rep ; 7(1): 12758, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28986582

ABSTRACT

Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) are key metabolic enzymes that are mutated in a variety of cancers to confer a gain-of-function activity resulting in the accumulation of an oncometabolite, D-2-hydroxyglutarate (2-HG). Accumulation of 2-HG can result in epigenetic dysregulation and a block in cellular differentiation, suggesting these mutations play a role in neoplasia. Based on its potential as a cancer target, a number of small molecule inhibitors have been developed to specifically inhibit mutant forms of IDH (mIDH1 and mIDH2). We present a comprehensive suite of in vitro preclinical drug development assays that can be used as a tool-box to identify lead compounds for mIDH drug discovery programs, as well as what we believe is the most comprehensive publically available dataset on the top mIDH inhibitors. This involved biochemical, cell-based, and tier-one ADME techniques.


Subject(s)
Drug Discovery , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Cell Differentiation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Stability , Fluorescence , Glutarates/metabolism , High-Throughput Screening Assays , Histones/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Methylation , Models, Biological , Monocytes/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , THP-1 Cells
15.
PET Clin ; 12(2): 235-241, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28267456

ABSTRACT

Prostate cancer (PCa) is the most common noncutaneous malignancy diagnosed in men. Despite the large number of men who will suffer from PCa at some point during their lives, conventional imaging modalities for this important disease (contrast-enhanced computed tomography, bone scan, and MR imaging) have provided only marginal to moderate success in appropriately guiding patient management in certain clinical contexts. In this review, the authors discuss radiofluorinated small molecule radiotracers that have been developed to bind to the transmembrane glycoprotein prostate-specific membrane antigen, a target that is nearly universally overexpressed on PCa epithelial cells.


Subject(s)
Fluorine Radioisotopes , Glutamate Carboxypeptidase II/antagonists & inhibitors , Prostatic Neoplasms/diagnostic imaging , Radiopharmaceuticals , Antigens, Surface , Cysteine/analogs & derivatives , Drug Evaluation, Preclinical/methods , Glutarates , Humans , Lysine/analogs & derivatives , Male , Organophosphonates , Positron Emission Tomography Computed Tomography/methods , Urea/analogs & derivatives
16.
Leukemia ; 31(10): 2020-2028, 2017 10.
Article in English | MEDLINE | ID: mdl-28232670

ABSTRACT

Neomorphic mutations in isocitrate dehydrogenase 1 (IDH1) are frequently found in several human cancer types including acute myeloid leukemia (AML) and lead to the production of high levels of the oncometabolite (R)-2-hydroxyglutarate (R-2HG). Here we report the characterization of BAY1436032, a novel pan-mutant IDH1 inhibitor, both in vitro and in vivo. BAY1436032 specifically inhibits R-2HG production and colony growth, and induces myeloid differentiation of AML cells carrying IDH1R132H, IDH1R132C, IDH1R132G, IDH1R132L and IDH1R132S mutations. In addition, the compound impacts on DNA methylation and attenuates histone hypermethylation. Oral administration of BAY1436032 led to leukemic blast clearance, myeloid differentiation, depletion of leukemic stem cells and prolonged survival in two independent patient-derived xenograft IDH1 mutant AML mouse models. Together, BAY1436032 is highly effective against all major types of IDH1 mutant AML.


Subject(s)
Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Enzyme Inhibitors/therapeutic use , Isocitrate Dehydrogenase/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Cell Line, Tumor , DNA Methylation/drug effects , Enzyme Inhibitors/pharmacology , Glutarates/metabolism , Histone Code/drug effects , Humans , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Methylation/drug effects , Mice , Molecular Targeted Therapy , Mutation , Mutation, Missense , Myeloid Cells/drug effects , Myelopoiesis/drug effects , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Point Mutation , Protein Processing, Post-Translational/drug effects , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
17.
Acta Neuropathol ; 133(4): 629-644, 2017 04.
Article in English | MEDLINE | ID: mdl-28124097

ABSTRACT

Mutations in codon 132 of isocitrate dehydrogenase (IDH) 1 are frequent in diffuse glioma, acute myeloid leukemia, chondrosarcoma and intrahepatic cholangiocarcinoma. These mutations result in a neomorphic enzyme specificity which leads to a dramatic increase of intracellular D-2-hydroxyglutarate (2-HG) in tumor cells. Therefore, mutant IDH1 protein is a highly attractive target for inhibitory drugs. Here, we describe the development and properties of BAY 1436032, a pan-inhibitor of IDH1 protein with different codon 132 mutations. BAY 1436032 strongly reduces 2-HG levels in cells carrying IDH1-R132H, -R132C, -R132G, -R132S and -R132L mutations. Cells not carrying IDH mutations were unaffected. BAY 1436032 did not exhibit toxicity in vitro or in vivo. The pharmacokinetic properties of BAY 1436032 allow for oral administration. In two independent experiments, BAY 1436032 has been shown to significantly prolong survival of mice intracerebrally transplanted with human astrocytoma carrying the IDH1R132H mutation. In conclusion, we developed a pan-inhibitor targeting tumors with different IDH1R132 mutations.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Astrocytoma/drug therapy , Benzimidazoles/pharmacology , Brain Neoplasms/drug therapy , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Aniline Compounds/chemistry , Aniline Compounds/pharmacokinetics , Aniline Compounds/toxicity , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Astrocytoma/enzymology , Astrocytoma/genetics , Benzimidazoles/chemistry , Benzimidazoles/pharmacokinetics , Benzimidazoles/toxicity , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Escherichia coli , Female , Glutarates/metabolism , HEK293 Cells , Humans , Isocitrate Dehydrogenase/metabolism , Mice, Inbred BALB C , Mice, Nude , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sarcoma/drug therapy , Sarcoma/enzymology , Sarcoma/genetics , Sf9 Cells , Xenograft Model Antitumor Assays
18.
J Inherit Metab Dis ; 40(1): 75-101, 2017 01.
Article in English | MEDLINE | ID: mdl-27853989

ABSTRACT

Glutaric aciduria type I (GA-I; synonym, glutaric acidemia type I) is a rare inherited metabolic disease caused by deficiency of glutaryl-CoA dehydrogenase located in the catabolic pathways of L-lysine, L-hydroxylysine, and L-tryptophan. The enzymatic defect results in elevated concentrations of glutaric acid, 3-hydroxyglutaric acid, glutaconic acid, and glutaryl carnitine in body tissues, which can be reliably detected by gas chromatography/mass spectrometry (organic acids) and tandem mass spectrometry (acylcarnitines). Most untreated individuals with GA-I experience acute encephalopathic crises during the first 6 years of life that are triggered by infectious diseases, febrile reaction to vaccinations, and surgery. These crises result in striatal injury and consequent dystonic movement disorder; thus, significant mortality and morbidity results. In some patients, neurologic disease may also develop without clinically apparent crises at any age. Neonatal screening for GA-I us being used in a growing number of countries worldwide and is cost effective. Metabolic treatment, consisting of low lysine diet, carnitine supplementation, and intensified emergency treatment during catabolism, is effective treatment and improves neurologic outcome in those individuals diagnosed early; treatment after symptom onset, however, is less effective. Dietary treatment is relaxed after age 6 years and should be supervised by specialized metabolic centers. The major aim of this second revision of proposed recommendations is to re-evaluate the previous recommendations (Kölker et al. J Inherit Metab Dis 30:5-22, 2007b; J Inherit Metab Dis 34:677-694, 2011) and add new research findings, relevant clinical aspects, and the perspective of affected individuals.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/drug therapy , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/drug therapy , Glutaryl-CoA Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic/metabolism , Dietary Supplements , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Lysine/metabolism
19.
Int J Hematol ; 104(4): 491-501, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27380194

ABSTRACT

Iron overload remains a concern in myelodysplastic syndrome (MDS) patients. Iron chelation therapy (ICT) thus plays an integral role in the management of these patients. Moreover, ICT has been shown to prolong leukemia-free survival in MDS patients; however, the mechanisms responsible for this effect are unclear. Iron is a key molecule for regulating cytosolic aconitase 1 (ACO1). Additionally, the mutation of isocitrate dehydrogenase (IDH), the enzyme downstream of ACO1 in the TCA cycle, is associated with epigenetic abnormalities secondary to 2-hydroxyglutarate (2-HG) and DNA methylation. However, epigenetic abnormalities observed in many MDS patients occur without IDH mutation. We hypothesized that iron itself activates the ACO1-IDH pathway, which may increase 2-HG and DNA methylation, and eventually contribute to leukemogenesis without IDH mutation. Using whole RNA sequencing of bone marrow cells in iron-overloaded mice, we observed that the enzymes, phosphoglucomutase 1, glycogen debranching enzyme, and isocitrate dehydrogenase 1 (Idh1), which are involved in glycogen and glucose metabolism, were increased. Digital PCR further showed that Idh1 and Aco1, enzymes involved in the TCA cycle, were also elevated. Additionally, enzymatic activities of TCA cycle and methylated DNA were increased. Iron chelation reversed these phenomena. In conclusion, iron activation of glucose metabolism causes an increase of 2-HG and DNA methylation.


Subject(s)
Bone Marrow/metabolism , DNA Methylation/drug effects , Iron Regulatory Protein 1/metabolism , Iron/pharmacology , Isocitrate Dehydrogenase/metabolism , Animals , Carcinogenesis/chemically induced , Glucose/metabolism , Glutarates/blood , Iron Regulatory Protein 1/drug effects , Isocitrate Dehydrogenase/drug effects , Mice
20.
J Lipid Res ; 57(8): 1455-64, 2016 08.
Article in English | MEDLINE | ID: mdl-27313057

ABSTRACT

Statins are competitive inhibitors of HMG-CoA reductase, the rate-limiting enzyme of cholesterol synthesis. Statins reduce plasma cholesterol levels, but whether this is actually caused by inhibition of de novo cholesterol synthesis has not been clearly established. Using three different statins, we investigated the effects on cholesterol metabolism in mice in detail. Surprisingly, direct measurement of whole body cholesterol synthesis revealed that cholesterol synthesis was robustly increased in statin-treated mice. Measurement of organ-specific cholesterol synthesis demonstrated that the liver is predominantly responsible for the increase in cholesterol synthesis. Excess synthesized cholesterol did not accumulate in the plasma, as plasma cholesterol decreased. However, statin treatment led to an increase in cholesterol removal via the feces. Interestingly, enhanced cholesterol excretion in response to rosuvastatin and lovastatin treatment was mainly mediated via biliary cholesterol secretion, whereas atorvastatin mainly stimulated cholesterol removal via the transintestinal cholesterol excretion pathway. Moreover, we show that plasma cholesterol precursor levels do not reflect cholesterol synthesis rates during statin treatment in mice. In conclusion, cholesterol synthesis is paradoxically increased upon statin treatment in mice. However, statins potently stimulate the excretion of cholesterol from the body, which sheds new light on possible mechanisms underlying the cholesterol-lowering effects of statins.


Subject(s)
Cholesterol/biosynthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver/metabolism , Lovastatin/pharmacology , Animals , Cholesterol/blood , Drug Evaluation, Preclinical , Gene Expression/drug effects , Glutarates/metabolism , Hypercholesterolemia/blood , Hypercholesterolemia/drug therapy , Intestinal Elimination/drug effects , Intestine, Small/drug effects , Intestine, Small/metabolism , Liver/drug effects , Male , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL