Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Epilepsy Behav ; 146: 109363, 2023 09.
Article in English | MEDLINE | ID: mdl-37499576

ABSTRACT

Lysine, as an essential amino acid, predominantly undergoes metabolic processes through the saccharopine pathway, whereas a smaller fraction follows the pipecolic acid pathway. Although the liver is considered the primary organ for lysine metabolism, it is worth noting that lysine catabolism also takes place in other tissues and organs throughout the body, including the brain. Enzyme deficiency caused by pathogenic variants in its metabolic pathway may lead to a series of neurometabolic diseases, among which glutaric aciduria type 1 and pyridoxine-dependent epilepsy have the most significant clinical manifestations. At present, through research, we have a deeper understanding of the multiple pathophysiological mechanisms related to these diseases, including intracerebral accumulation of neurotoxic metabolites, imbalance between GABAergic and glutamatergic neurotransmission, energy deprivation due to metabolites, and the dysfunction of antiquitin. Because of the complexity of these diseases, their clinical manifestations are also diverse. The early implementation of lysine-restricted diets and supplementation with arginine and carnitine has reported positive impacts on the neurodevelopmental outcomes of patients. Presently, there is more robust evidence supporting the effectiveness of these treatments in glutaric aciduria type 1 compared with pyridoxine-dependent epilepsy.


Subject(s)
Brain Diseases, Metabolic , Epilepsy , Humans , Lysine/metabolism , Epilepsy/metabolism , Brain Diseases, Metabolic/complications , Brain Diseases, Metabolic/metabolism
2.
J Child Neurol ; 38(6-7): 446-453, 2023 05.
Article in English | MEDLINE | ID: mdl-37128731

ABSTRACT

PURPOSE: To assess the diagnostic value of the thalamus L-sign on magnetic resonance imaging (MRI) in distinguishing between periventricular leukomalacia and neurometabolic disorders in pediatric patients. METHODS: In this retrospective study, clinical and imaging information was collected from 50 children with periventricular leukomalacia and 52 children with neurometabolic disorders. MRI was used to evaluate the L-sign of the thalamus (ie, injury to the posterolateral thalamus) and the lobar distribution of signal intensity changes. Age, sex, gestational age, and level of Gross Motor Function Classification System (only for periventricular leukomalacia) constituted the clinical parameters. Statistical evaluation of group differences for imaging and clinical variables were conducted using univariable statistical methods. The intra- and inter-observer agreement was evaluated using Cohen's kappa. Univariable or multivariable logistic regression was employed for selection of variables, determining independent predictors, and modeling. RESULTS: The thalamus L-sign was observed in 70% (35/50) of patients in the periventricular leukomalacia group, but in none of the patients with neurometabolic disorder (P < .001). The gestational age between groups varied significantly (P < .001). Involvement of frontal, parietal, and occipital lobes differed significantly between groups (P < .001). In the logistic regression, the best model included negative thalamus L-sign and gestational age, yielding an area under the curve, accuracy, sensitivity, specificity, and precision values of 0.995, 96.1%, 96%, 96.2%, and 96%, respectively. Both the lack of thalamus L-sign and gestational age were independent predictors (P < .001). CONCLUSIONS: The thalamus L-sign and gestational age may be useful in distinguishing between periventricular leukomalacia and neurometabolic disorders.


Subject(s)
Brain Diseases, Metabolic , Leukomalacia, Periventricular , Thalamus , Child , Humans , Brain Diseases, Metabolic/diagnostic imaging , Brain Diseases, Metabolic/pathology , Diagnosis, Differential , Frontal Lobe , Gestational Age , Infant, Premature , Leukomalacia, Periventricular/diagnostic imaging , Leukomalacia, Periventricular/pathology , Logistic Models , Magnetic Resonance Imaging , Occipital Lobe , Parietal Lobe , Retrospective Studies , Thalamus/diagnostic imaging , Thalamus/injuries , Thalamus/pathology , Biomarkers , Motor Skills , Male , Female , Infant , Child, Preschool , Adolescent
3.
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
4.
AJNR Am J Neuroradiol ; 43(6): 919-925, 2022 06.
Article in English | MEDLINE | ID: mdl-35589136

ABSTRACT

BACKGROUND AND PURPOSE: Considerable overlap exists in the MR imaging features of hypoglycemic injury and hypoxic-ischemic brain injury, with similar predilections for the occipital and parietal lobes. In partial, prolonged hypoxia-ischemia, there is cortical destruction at the interarterial watershed zones, and in concomitant hypoglycemia and hypoxia-ischemia, an exaggerated final common pathway injury occurs. We interrogated secondary white matter tract-based thalamic injury as a tool to separate pure injuries in each group. MATERIALS AND METHODS: A retrospective observational study of the MRIs of 320 children with a history of hypoxia-ischemia and/or hypoglycemia was undertaken with 3 major subgroups: 1) watershed-type hypoxic-ischemic injury, 2) neonatal hypoglycemia, and 3) both perinatal hypoxia-ischemia and proved hypoglycemia. Cerebral and thalamic injuries were assessed, particularly hyperintensity of the posterolateral margin of the thalami. A modified Poisson regression model was used to assess factors associated with such thalamic injury. RESULTS: Parieto-occipital injuries occurred commonly in patients with hypoglycemia and/or hypoxia-ischemia. Eighty-five of 99 (86%) patients with partial, prolonged hypoxia-ischemia exhibited the thalamus L-sign. This sign was also observed in patients who had both hypoglycemia and hypoxia-ischemia, predominantly attributable to the latter. Notably, the risk of a thalamus L-sign injury was 2.79 times higher when both the parietal and occipital lobes were injured compared with when they were not involved (95% CI, 1.25-6.23; P = .012). The thalamus L-sign was not depicted in patients with pure hypoglycemia. CONCLUSIONS: We propose the thalamus L-sign as a biomarker of partial, prolonged hypoxia-ischemia, which is exaggerated in combined hypoglycemic/hypoxic-ischemic injury.


Subject(s)
Brain Diseases, Metabolic , Brain Injuries , Hypoglycemia , Hypoxia-Ischemia, Brain , Biomarkers , Brain , Child , Female , Humans , Hypoglycemia/complications , Hypoglycemic Agents , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/diagnostic imaging , Infant, Newborn , Pregnancy , Thalamus/diagnostic imaging
5.
Oxid Med Cell Longev ; 2022: 8295580, 2022.
Article in English | MEDLINE | ID: mdl-35087621

ABSTRACT

This study was designed to explore the potential mechanisms of electroacupuncture (EA) in treating inflammatory bowel disease- (IBD-) related anxiety and mood disorders. A colitis model was induced in rats with 2, 4, 6-trinitrohydrosulfonic acid (TNBS), followed by ST36 and SP6 targeted therapy by EA or sham EA treatment. The elevated plus maze (EPM) and open-field test (OFT) were performed to assess the state of anxiety and depression-like behavior. Tests were carried out by 16S rDNA amplification sequence, 1H nuclear magnetic resonance (1H NMR) spectroscopy, immunofluorescence staining, and enzyme-linked immunosorbent assay (ELISA). The analyses detailed metabolic alterations and the Toll-like receptor 4 (TLR4) signaling pathway/NOD-like receptor protein 3 (NLRP3) inflammasome in rats' hippocampal region. Furthermore, the activity of the hypothalamic-pituitary adrenal (HPA) axis and gut microbiome was assessed. As a result of treatment, EA significantly improved in the behavioral tests and altered the composition of the gut microbiome through a significant increase in the density of short chain fatty acids (SCFAs) producers mainly including Ruminococcaceae, Phascolarctobacterium, and Akkermansiaceae. EA upregulated the metabolites of the hippocampus mainly containing l-glutamine and gamma-aminobutyric acid (GABA), as well as ZO-1 expression. Whereas the treatment blocked the TLR4/nuclear factor- kappa B (NF-κB) signaling pathways and NLRP3 inflammasomes, along with downregulating the interleukin- (IL-) 1ß level. The hyperactivity of the HPA axis was also diminished. In conclusion, EA at ST36 and SP6 attenuated anxiety and depression-like behavior in colitis model rats through their effects on the gut microbiome by modulating the hippocampal inflammatory response and metabolic disorders, as well as the HPA axis. This study provides evidence for clinical application of EA to serve as an adjunctive treatment for IBD-related anxiety and depression.


Subject(s)
Anxiety/therapy , Brain Diseases, Metabolic/physiopathology , Depression/therapy , Electroacupuncture/methods , Hippocampus/physiopathology , Inflammation/physiopathology , Inflammatory Bowel Diseases/therapy , Animals , Male , Rats , Rats, Sprague-Dawley
6.
Arch Biochem Biophys ; 709: 108970, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34181873

ABSTRACT

Glutaric acidemia type 1 (GA1) is caused by glutaryl-CoA dehydrogenase deficiency that leads to a blockage in the metabolic route of the amino acids lysine and tryptophan and subsequent accumulation of glutaric acid (GA), 3-hydroxyglutaric acids and glutarylcarnitine (C5DC). Patients predominantly manifest neurological symptoms, associated with acute striatal degeneration, as well as progressive cortical and striatum injury whose pathogenesis is not yet fully established. Current treatment includes protein/lysine restriction and l-carnitine supplementation of (L-car). The aim of this work was to evaluate behavior parameters and pro-inflammatory factors (cytokines IL-1ß, TNF-α and cathepsin-D levels), as well as the anti-inflammatory cytokine IL10 in striatum of knockout mice (Gcdh-/-) and wild type (WT) mice submitted to a normal or a high Lys diet. The potential protective effects of L-car treatment on these parameters were also evaluated. Gcdh-/- mice showed behavioral changes, including lower motor activity (decreased number of crossings) and exploratory activity (reduced number of rearings). Also, Gcdh-/- mice had significantly higher concentrations of glutarylcarnitine (C5DC) in blood and cathepsin-D (CATD), interleukin IL-1ß and tumor factor necrosis alpha (TNF-α) in striatum than WT mice. Noteworthy, L-car treatment prevented most behavioral alterations, normalized CATD levels and attenuated IL-1ß levels in striatum of Gcdh-/- mice. Finally, IL-1ß was positively correlated with CATD and C5DC levels and L-car was negatively correlated with CATD. Our results demonstrate behavioral changes and a pro-inflammatory status in striatum of the animal model of GA1 and, most importantly, L-car showed important protective effects on these alterations.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Brain Diseases, Metabolic/drug therapy , Carnitine/therapeutic use , Glutaryl-CoA Dehydrogenase/deficiency , Inflammation/drug therapy , Neuroprotective Agents/therapeutic use , Amino Acid Metabolism, Inborn Errors/genetics , Animals , Brain Diseases, Metabolic/genetics , Carnitine/analogs & derivatives , Carnitine/metabolism , Cathepsin D/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Glutaryl-CoA Dehydrogenase/genetics , Grooming/drug effects , Inflammation/genetics , Interleukin-1beta/metabolism , Locomotion/drug effects , Lysine/pharmacology , Mice, Knockout , Open Field Test/drug effects , Transforming Growth Factor beta/metabolism
7.
Nat Commun ; 11(1): 5906, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33219201

ABSTRACT

Hypothalamic inflammation plays an important role in disrupting feeding behavior and energy homeostasis as well as in the pathogenesis of obesity and diabetes. Here, we show that pyruvate dehydrogenase kinase (PDK)-2 plays a role in hypothalamic inflammation and its sequelae in mouse models of diabetes. Cell type-specific genetic ablation and pharmacological inhibition of PDK2 in hypothalamic astrocytes suggest that hypothalamic astrocytes are involved in the diabetic phenotype. We also show that the PDK2-lactic acid axis plays a regulatory role in the observed metabolic imbalance and hypothalamic inflammation in mouse primary astrocyte and organotypic cultures, through the AMPK signaling pathway and neuropeptidergic circuitry governing feeding behavior. Our findings reveal that PDK2 ablation or inhibition in mouse astrocytes attenuates diabetes-induced hypothalamic inflammation and subsequent alterations in feeding behavior.


Subject(s)
Astrocytes/metabolism , Diabetes Mellitus/metabolism , Hypothalamus , Inflammation/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Animals , Brain Diseases, Metabolic , Disease Models, Animal , Feeding Behavior , Hypothalamus/cytology , Hypothalamus/metabolism , Hypothalamus/pathology , Mice , Obesity , Signal Transduction
8.
Mol Genet Metab ; 131(3): 325-340, 2020 11.
Article in English | MEDLINE | ID: mdl-33069577

ABSTRACT

Glutaric acidemia type 1 (GA1) is a disorder of cerebral organic acid metabolism resulting from biallelic mutations of GCDH. Without treatment, GA1 causes striatal degeneration in >80% of affected children before two years of age. We analyzed clinical, biochemical, and developmental outcomes for 168 genotypically diverse GA1 patients managed at a single center over 31 years, here separated into three treatment cohorts: children in Cohort I (n = 60; DOB 2006-2019) were identified by newborn screening (NBS) and treated prospectively using a standardized protocol that included a lysine-free, arginine-enriched metabolic formula, enteral l-carnitine (100 mg/kg•day), and emergency intravenous (IV) infusions of dextrose, saline, and l-carnitine during illnesses; children in Cohort II (n = 57; DOB 1989-2018) were identified by NBS and treated with natural protein restriction (1.0-1.3 g/kg•day) and emergency IV infusions; children in Cohort III (n = 51; DOB 1973-2016) did not receive NBS or special diet. The incidence of striatal degeneration in Cohorts I, II, and III was 7%, 47%, and 90%, respectively (p < .0001). No neurologic injuries occurred after 19 months of age. Among uninjured children followed prospectively from birth (Cohort I), measures of growth, nutritional sufficiency, motor development, and cognitive function were normal. Adherence to metabolic formula and l-carnitine supplementation in Cohort I declined to 12% and 32%, respectively, by age 7 years. Cessation of strict dietary therapy altered plasma amino acid and carnitine concentrations but resulted in no serious adverse outcomes. In conclusion, neonatal diagnosis of GA1 coupled to management with lysine-free, arginine-enriched metabolic formula and emergency IV infusions during the first two years of life is safe and effective, preventing more than 90% of striatal injuries while supporting normal growth and psychomotor development. The need for dietary interventions and emergency IV therapies beyond early childhood is uncertain.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/genetics , Brain/metabolism , Corpus Striatum/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , Amino Acid Metabolism, Inborn Errors/diet therapy , Amino Acid Metabolism, Inborn Errors/epidemiology , Amino Acid Metabolism, Inborn Errors/metabolism , Brain/pathology , Brain Diseases, Metabolic/diet therapy , Brain Diseases, Metabolic/epidemiology , Brain Diseases, Metabolic/metabolism , Carnitine/metabolism , Child , Child, Preschool , Corpus Striatum/pathology , Diet , Female , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Infant , Infant, Newborn , Lysine/metabolism , Male
9.
Eur Rev Med Pharmacol Sci ; 24(19): 10051-10053, 2020 10.
Article in English | MEDLINE | ID: mdl-33090410

ABSTRACT

OBJECTIVE: Long-term survival of patients with neonatal-onset carbamoyl-phosphate synthetase 1 deficiency (CPS1D), an autosomal recessive disorder characterized by repeated, life-threatening hyperammonemia, is rare. We describe the diagnosis and clinical management of a teenager with neonatal-onset CPS1D who did not undergo therapeutic liver transplantation. CASE REPORT: Following emergent neonatal therapy, the patient was diagnosed with CPS1D based on clinical, radiological, biochemical and genetic analyses. Her clinical course, neurobehavioral development and therapeutic interventions are presented and discussed. RESULTS: Born from nonconsanguineous parents, the proband underwent phototherapy for neonatal jaundice, associated with acute encephalopathy, apnea and cerebral edema. Based on blood and urinary biochemical abnormalities, neonatal-onset CPS1D was diagnosed. Her hyperammonemia was corrected by hemodialysis, followed by sodium benzoate, L-arginine, levocarnitine and protein-free diet therapy. Because of a relapse and persistent neurobehavioral regression by age 1, a planned liver transplantation was cancelled. At age 10, sodium phenylbutyrate was substituted as ammonia scavenger. Genetic testing revealed compound heterozygote c.2359C>T (R787X) and c.236+6T>C variants of CPS1, confirming her diagnosis. Despite severe neurological sequelae, the patient is 16 and in stable condition. CONCLUSIONS: Our case suggests that early hemodialysis and pharmacologic interventions for acute neonatal hyperammonemia can improve the prognosis of patients with neonatal-onset CPS1D.


Subject(s)
Arginine/therapeutic use , Brain Diseases, Metabolic/therapy , Carbamoyl-Phosphate Synthase I Deficiency Disease/therapy , Carnitine/therapeutic use , Hyperammonemia/therapy , Phenylbutyrates/therapeutic use , Renal Dialysis , Sodium Benzoate/therapeutic use , Female , Humans , Infant, Newborn
10.
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
11.
Nutrients ; 12(10)2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33081139

ABSTRACT

Glutaric aciduria type 1 (GA-1) is a cerebral organic aciduria characterized by striatal injury and progressive movement disorder. Nutrition management shifted from a general restriction of intact protein to targeted restriction of lysine and tryptophan. Recent guidelines advocate for a low-lysine diet using lysine-free, tryptophan-reduced medical foods. GA-1 guideline recommendations for dietary management of patients over the age of six are unclear, ranging from avoiding excessive intake of intact protein to counting milligrams of lysine intake. A 22-question survey on the nutrition management of GA-1 was developed with the goal of understanding approaches to diet management for patients identified by newborn screening under age six years compared to management after diet liberalization, as well as to gain insight into how clinicians define diet liberalization. Seventy-six responses (25% of possible responses) to the survey were received. Nutrition management with GA-1 is divergent among surveyed clinicians. There was congruency among survey responses to the guidelines, but there is still uncertainty about how to counsel patients on diet optimization and when diet liberalization should occur. Ongoing clinical research and better understanding of the natural history of this disease will help establish stronger recommendations from which clinicians can best counsel families.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diet therapy , Brain Diseases, Metabolic/diet therapy , Child Nutritional Physiological Phenomena/physiology , Diet Therapy/methods , Dietary Proteins/administration & dosage , Glutaryl-CoA Dehydrogenase/deficiency , Infant Nutritional Physiological Phenomena/physiology , Lysine/adverse effects , Tryptophan/adverse effects , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Practice Guidelines as Topic , Recommended Dietary Allowances , Surveys and Questionnaires
12.
Int J Mol Sci ; 21(19)2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32992790

ABSTRACT

Riboflavin is the biological precursor of two important flavin cofactors-flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN)-that are critical prosthetic groups in several redox enzymes. While dietary supplementation with riboflavin is a recognized support therapy in several inborn errors of metabolism, it has yet unproven benefits in several other pathologies affecting flavoproteins. This is the case for glutaric aciduria type I (GA-I), a rare neurometabolic disorder associated with mutations in the GCDH gene, which encodes for glutaryl-coenzyme A (CoA) dehydrogenase (GCDH). Although there are a few reported clinical cases that have responded to riboflavin intake, there is still not enough molecular evidence supporting therapeutic recommendation. Hence, it is necessary to elucidate the molecular basis in favor of riboflavin supplementation in GA-I patients. Here, using a combination of biochemical and biophysical methodologies, we investigate the clinical variant GCDH-p.Val400Met as a model for a phenotype associated with severe deflavinylation. Through a systematic analysis, we establish that recombinant human GCDH-p.Val400Met is expressed in a nonfunctional apo form, which is mainly monomeric rather than tetrameric. However, we show that exogenous FAD is a driver for structural reorganization of the mutant enzyme with concomitant functional recovery, improved thermolability, and resistance to trypsin digestion. Overall, these results establish proof of principle for the beneficial effects of riboflavin supplementation in GA-I patients.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Brain Diseases, Metabolic , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , Riboflavin , Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic/metabolism , Glutaryl-CoA Dehydrogenase/chemistry , Glutaryl-CoA Dehydrogenase/drug effects , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Mutation , Protein Folding/drug effects , Protein Stability/drug effects , Recombinant Proteins , Riboflavin/pharmacology
13.
Genes (Basel) ; 11(8)2020 08 10.
Article in English | MEDLINE | ID: mdl-32785089

ABSTRACT

Monogenic metabolic disorders of hepatic origin number in the hundreds, and for many, liver transplantation remains the only cure. Liver-targeted gene therapy is an attractive treatment modality for many of these conditions, and there have been significant advances at both the preclinical and clinical stages. Viral vectors, including retroviruses, lentiviruses, adenovirus-based vectors, adeno-associated viruses and simian virus 40, have differing safety, efficacy and immunogenic profiles, and several of these have been used in clinical trials with variable success. In this review, we profile viral vectors and non-viral vectors, together with various payloads, including emerging therapies based on RNA, that are entering clinical trials. Genome editing technologies are explored, from earlier to more recent novel approaches that are more efficient, specific and safe in reaching their target sites. The various curative approaches for the multitude of monogenic hepatic metabolic disorders currently at the clinical development stage portend a favorable outlook for this class of genetic disorders.


Subject(s)
Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/therapy , Genetic Therapy , Liver Diseases/genetics , Liver Diseases/therapy , Animals , Biomarkers , CRISPR-Cas Systems , Clinical Studies as Topic , Disease Management , Drug Evaluation, Preclinical , Gene Editing , Gene Expression , Gene Transfer Techniques , Genetic Predisposition to Disease , Genetic Therapy/methods , Genetic Vectors/classification , Genetic Vectors/genetics , Humans , Organ Specificity/genetics , Transduction, Genetic , Treatment Outcome
14.
BMJ Case Rep ; 12(11)2019 Nov 25.
Article in English | MEDLINE | ID: mdl-31772134

ABSTRACT

A 26-year-old man presented at the emergency department with confusion and decreased consciousness after several days of vomiting. In the preceding 6 months, he had used a 2-litre tank of nitrous oxide (N2O) weekly. His metabolic encephalopathy was caused by hyperammonaemia which probably resulted from interference of N2O-induced vitamin B12 deficiency with ammonia degradation. A catabolic state might have contributed to the hyperammonaemia in this case. After treatment with vitamin B12 and lactulose, both his consciousness and hyperammonaemia improved. He reported no residual complaints after 3 months of follow-up. Since N2O is increasingly used as a recreational drug, we recommend considering hyperammonaemia as a cause of metabolic encephalopathy in cases of N2O use and altered mental status.


Subject(s)
Brain Diseases, Metabolic/chemically induced , Confusion/diagnosis , Consciousness Disorders/diagnosis , Hyperammonemia/chemically induced , Nitrous Oxide/adverse effects , Adult , Brain Diseases, Metabolic/drug therapy , Confusion/etiology , Consciousness Disorders/etiology , Diagnosis, Differential , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/therapeutic use , Humans , Hyperammonemia/complications , Lactulose/administration & dosage , Lactulose/therapeutic use , Male , Treatment Outcome , Vitamin B 12/administration & dosage , Vitamin B 12/therapeutic use , Vitamin B 12 Deficiency/complications , Vitamin B Complex/administration & dosage , Vitamin B Complex/therapeutic use , Vomiting/diagnosis
15.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2420-2427, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31181292

ABSTRACT

The deficiency of the enzyme glutaryl-CoA dehydrogenase leads to predominant accumulation of glutaric acid (GA) in the organism and is known as glutaric acidemia type I (GA1). Despite the mechanisms of brain damage involved in GA1 are not fully understood, oxidative stress may be involved in this process. Treatment is based on protein/lysine (Lys) restriction and l-carnitine (L-car) supplementation. L-car was recently shown to have an important antioxidant role. A knockout mice model (Gcdh-/-) submitted to a dietary overload of Lys was developed to better understand the GA1 pathogenesis. In this study, we evaluated L-car and glutarylcarnitine levels, the lipid and protein damage, reactive oxygen species (ROS) production and antioxidant enzymes activities in striatum of Gcdh-/- and wild-type (WT) mice. We also determined the effect of the L-car treatment on these parameters. Thirty-day-old Gcdh-/- and WT mice were fed a normal chow (0.9% Lys) or submitted to a high Lys diet (4.7%) for 72 h. Additionally, these animals were administered with three intraperitoneal injections of saline or L-car in different times. Gcdh-/- mice were deficient in L-car and presented a higher glutarylcarnitine levels. They also presented lipid and protein damage, an increased ROS production and altered antioxidant enzymes compared to WT mice. Additionally, mice exposed to Lys overload presented higher alterations in these parameters than mice under normal diet, which were significantly decreased or normalized in those receiving L-car. Thus, we demonstrated a new beneficial effect of the L-car treatment attenuating or abolishing the oxidative stress process in Gcdh-/- mice.


Subject(s)
Carnitine/pharmacology , Corpus Striatum/metabolism , Glutaryl-CoA Dehydrogenase/genetics , Lysine/pharmacology , Oxidative Stress/drug effects , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Amino Acid Metabolism, Inborn Errors/veterinary , Animals , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/pathology , Brain Diseases, Metabolic/veterinary , Carnitine/analogs & derivatives , Carnitine/metabolism , Diet/veterinary , Disease Models, Animal , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/metabolism , Glutathione Peroxidase/metabolism , Lysine/blood , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
16.
Am Soc Clin Oncol Educ Book ; 38: 822-828, 2018 May 23.
Article in English | MEDLINE | ID: mdl-30231327

ABSTRACT

There are 15.5 million cancer survivors in the United States because of, in part, improvements in therapy. As a result, there will be an increased burden of long- and late-term complications of cancer care, such as metabolic alterations. These metabolic changes will include alterations in bone resorption, obesity, hypercholesterolemia, and diabetes mellitus. The majority of cancer treatment-related toxicities have focused on endocrine therapy; however, chemotherapy and supportive medications, such as steroids, contribute to the development of these disorders. Because of the chronicity of these metabolic changes and their impact on morbidity, cancer risk, and outcomes as well other negative effects, including musculoskeletal pain and vasomotor symptoms, alternative strategies must be developed. These strategies should include nonpharmacologic approaches. Here, we summarize metabolic changes secondary to cancer care and integrative approaches to help alleviate therapy-associated toxicities.


Subject(s)
Combined Modality Therapy/adverse effects , Neoplasms/complications , Androgen Antagonists/adverse effects , Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/therapeutic use , Brain Diseases, Metabolic/etiology , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/pathology , Combined Modality Therapy/methods , Delivery of Health Care, Integrated , Disease Management , Glucocorticoids/adverse effects , Glucocorticoids/therapeutic use , Humans , Hypogonadism/complications , Metabolic Syndrome/etiology , Neoplasms/metabolism , Neoplasms/therapy , Osteoporosis/etiology
17.
J Cell Biochem ; 119(12): 10021-10032, 2018 12.
Article in English | MEDLINE | ID: mdl-30129250

ABSTRACT

The deficiency of the enzyme glutaryl-CoA dehydrogenase, known as glutaric acidemia type I (GA-I), leads to the accumulation of glutaric acid (GA) and glutarilcarnitine (C5DC) in the tissues and body fluids, unleashing important neurotoxic effects. l-carnitine (l-car) is recommended for the treatment of GA-I, aiming to induce the excretion of toxic metabolites. l-car has also demonstrated an important role as antioxidant and anti-inflammatory in some neurometabolic diseases. This study evaluated GA-I patients at diagnosis moment and treated the oxidative damage to lipids, proteins, and the inflammatory profile, as well as in vivo and in vitro DNA damage, reactive nitrogen species (RNS), and antioxidant capacity, verifying if the actual treatment with l-car (100 mg kg-1 day-1 ) is able to protect the organism against these processes. Significant increases of GA and C5DC were observed in GA-I patients. A deficiency of carnitine in patients before the supplementation was found. GA-I patients presented significantly increased levels of isoprostanes, di-tyrosine, urinary oxidized guanine species, and the RNS, as well as a reduced antioxidant capacity. The l-car supplementation induced beneficial effects reducing these biomarkers levels and increasing the antioxidant capacity. GA, in three different concentrations, significantly induced DNA damage in vitro, and the l-car was able to prevent this damage. Significant increases of pro-inflammatory cytokines IL-6, IL-8, GM-CSF, and TNF-α were shown in patients. Thus, the beneficial effects of l-car presented in the treatment of GA-I are due not only by increasing the excretion of accumulated toxic metabolites, but also by preventing oxidative damage.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic/metabolism , Carnitine/pharmacology , DNA Damage , Glutaryl-CoA Dehydrogenase/deficiency , Oxidative Stress , Antioxidants/pharmacology , Antioxidants/therapeutic use , Carnitine/therapeutic use , Child , Child, Preschool , Female , Glutaryl-CoA Dehydrogenase/drug effects , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Infant , Male , Protective Agents/pharmacology , Protective Agents/therapeutic use , Reactive Nitrogen Species
18.
Article in Russian | MEDLINE | ID: mdl-29460912

ABSTRACT

The review presents current concepts about the problem of cerebral insulin resistance (IR). It has now been established that cerebral IR plays a key role in the pathogenesis of degenerative and metabolic diseases of the brain. Based on literature data and own clinical experience, the authors recommend to use the standardized extract of ginkgo biloba EGb761 as a cellular protector, which increases insulin sensitivity of cells and reduces atherogenesis, in order to improve cognitive functions and quality of life in patients with diabetes mellitus.


Subject(s)
Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/etiology , Diabetes Mellitus, Type 2/complications , Insulin Resistance , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Animals , Atherosclerosis/complications , Atherosclerosis/drug therapy , Cognition/drug effects , Ginkgo biloba , Humans , Mice , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Quality of Life
19.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 263-273, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29107091

ABSTRACT

Alzheimer's disease (AD) is an amyloid-related neurodegenerative disorder and is also considered to be a metabolic disease. Thus, investigation of metabolic mechanisms of amyloid pathology progression is of substantial importance for the diagnosis, prevention and treatment of AD. In the present study, cognitive function and brain metabolism were explored in the transgenic APP/PS1 mouse model of amyloid pathology at different ages. Using an NMR-based metabolomic approach, we examined metabolic changes in six different brain regions of wild-type and APP/PS1 mice at 1, 5 and 10months of age. Learning and memory performance in mice was evaluated using the Morris water maze test. Furthermore, a generalized linear mixed model was employed to analyze the interaction effect between the mouse-type and brain region (or age) on metabolic alterations. Brain region-specific changes in energy metabolism occurred prior to a very early-stage of amyloid pathology (1month of age) in APP/PS1 mice. A hypermetabolic state was identified in the brains of APP/PS1 mice at 5months of age, and the hypothalamus was identified as the main brain region that underwent significant metabolic alterations. The cognitive function of APP/PS1 mice was impaired at 10months of age; moreover, the hypermetabolic state identified in various brain regions at 5months of age was also significantly decreased. In conclusion, our results suggest that a hypothalamic metabolism abnormality may comprise a potential indicator for the early-diagnosis and monitoring of amyloid pathology progression.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Hypothalamus/metabolism , Hypothalamus/pathology , Presenilin-1/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/genetics , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Brain/metabolism , Brain/pathology , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/pathology , Cognition/physiology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/metabolism
20.
Sci Rep ; 7: 41032, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28102362

ABSTRACT

The neurologic manifestations of neonatal hyperbilirubinemia in the central nervous system (CNS) exhibit high variations in the severity and appearance of motor, auditory and cognitive symptoms, which is suggestive of a still unexplained selective topography of bilirubin-induced damage. By applying the organotypic brain culture (OBC: preserving in vitro the cellular complexity, connection and architecture of the in vivo brain) technique to study hyperbilirubinemia, we mapped the regional target of bilirubin-induced damage, demonstrated a multifactorial toxic action of bilirubin, and used this information to evaluate the efficacy of drugs applicable to newborns to protect the brain. OBCs from 8-day-old rat pups showed a 2-13 fold higher sensitivity to bilirubin damage than 2-day-old preparations. The hippocampus, inferior colliculus and cerebral cortex were the only brain regions affected, presenting a mixed inflammatory-oxidative mechanism. Glutamate excitotoxicity was appreciable in only the hippocampus and inferior colliculus. Single drug treatment (indomethacin, curcumin, MgCl2) significantly improved cell viability in all regions, while the combined (cocktail) administration of the three drugs almost completely prevented damage in the most affected area (hippocampus). Our data may supports an innovative (complementary to phototherapy) approach for directly protecting the newborn brain from bilirubin neurotoxicity.


Subject(s)
Bilirubin/toxicity , Brain Diseases, Metabolic/pathology , Brain/drug effects , Brain/pathology , Encephalitis/pathology , Hyperbilirubinemia/complications , Animals , Cell Survival/drug effects , Curcumin/pharmacology , Indomethacin/pharmacology , Inflammation/pathology , Magnesium Chloride/pharmacology , Models, Biological , Neuroprotective Agents/pharmacology , Organ Culture Techniques , Oxidative Stress , Rats
SELECTION OF CITATIONS
SEARCH DETAIL