Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Sci Rep ; 14(1): 11222, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38755170

ABSTRACT

Homocysteine (Hcy) and Hcy-thiolactone (HTL) affect fibrin clot properties and are linked to cardiovascular disease. Factors that influence fibrin clot properties and stroke are not fully understood. To study sulfur-containing amino acid metabolites, fibrin clot lysis time (CLT) and maximum absorbance (Absmax) in relation to stroke, we analyzed plasma and urine from 191 stroke patients (45.0% women, age 68 ± 12 years) and 291 healthy individuals (59.7% women, age 50 ± 17 years). Plasma and urinary levels of sulfur-containing amino acid metabolites and fibrin clot properties were significantly different in stroke patients compared to healthy individuals. Fibrin CLT correlated with fibrin Absmax in healthy males (R2 = 0.439, P = 0.000), females (R2 = 0.245, P = 0.000), female stroke patients (R2 = 0.187, P = 0.000), but not in male stroke patients (R2 = 0.008, P = ns). Fibrin CLT correlated with age in healthy females but not males while fibrin Absmax correlated with age in both sexes; these correlations were absent in stroke patients. In multiple regression analysis in stroke patients, plasma (p)CysGly, pMet, and MTHFR A1298C polymorphism were associated with fibrin Absmax, while urinary (u)HTL, uCysGly, and pCysGly were significantly associated with fibrin CLT. In healthy individuals, uHTL and uGSH were significantly associated with fibrin Absmax, while pGSH, and CBS T833C 844ins68 polymorphism were associated with fibrin CLT. In logistic regression, uHTL, uHcy, pCysGly, pGSH, MTHFR C677T polymorphism, and Absmax were independently associated with stroke. Our findings suggest that HTL and other sulfur-containing amino acid metabolites influence fibrin clot properties and the risk of stroke.


Subject(s)
Fibrin , Homocysteine , Ischemic Stroke , Humans , Male , Female , Homocysteine/blood , Homocysteine/analogs & derivatives , Homocysteine/metabolism , Homocysteine/urine , Aged , Middle Aged , Fibrin/metabolism , Ischemic Stroke/blood , Ischemic Stroke/metabolism , Ischemic Stroke/urine , Adult , Fibrin Clot Lysis Time , Risk Factors , Amino Acids, Sulfur/blood , Amino Acids, Sulfur/metabolism , Amino Acids, Sulfur/urine , Amino Acids/urine , Amino Acids/blood , Amino Acids/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Case-Control Studies , Aged, 80 and over , Stroke/metabolism , Stroke/blood , Stroke/urine
2.
Redox Biol ; 73: 103192, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38776754

ABSTRACT

BACKGROUND: In animals, dietary sulfur amino acid restriction (SAAR) improves metabolic health, possibly mediated by altering sulfur amino acid metabolism and enhanced anti-obesogenic processes in adipose tissue. AIM: To assess the effects of SAAR over time on the plasma and urine SAA-related metabolites (sulfurome) in humans with overweight and obesity, and explore whether such changes were associated with body weight, body fat and adipose tissue gene expression. METHODS: Fifty-nine subjects were randomly allocated to SAAR (∼2 g SAA, n = 31) or a control diet (∼5.6 g SAA, n = 28) consisting of plant-based whole-foods and supplemented with capsules to titrate contents of SAA. Sulfurome metabolites in plasma and urine at baseline, 4 and 8 weeks were measured using HPLC and LC-MS/MS. mRNA-sequencing of subcutaneous white adipose tissue (scWAT) was performed to assess changes in gene expression. Data were analyzed with mixed model regression. Principal component analyses (PCA) were performed on the sulfurome data to identify potential signatures characterizing the response to SAAR. RESULTS: SAAR led to marked decrease of the main urinary excretion product sulfate (p < 0.001) and plasma and/or 24-h urine concentrations of cystathionine, sulfite, thiosulfate, H2S, hypotaurine and taurine. PCA revealed a distinct metabolic signature related to decreased transsulfuration and H2S catabolism that predicted greater weight loss and android fat mass loss in SAAR vs. controls (all pinteraction < 0.05). This signature correlated positively with scWAT expression of genes in the tricarboxylic acid cycle, electron transport and ß-oxidation (FDR = 0.02). CONCLUSION: SAAR leads to distinct alterations of the plasma and urine sulfurome in humans, and predicted increased loss of weight and android fat mass, and adipose tissue lipolytic gene expression in scWAT. Our data suggest that SAA are linked to obesogenic processes and that SAAR may be useful for obesity and related disorders. TRIAL IDENTIFIER: https://clinicaltrials.gov/study/NCT04701346.


Subject(s)
Adipose Tissue , Amino Acids, Sulfur , Obesity , Overweight , Humans , Obesity/metabolism , Obesity/genetics , Male , Female , Overweight/metabolism , Overweight/genetics , Adult , Middle Aged , Adipose Tissue/metabolism , Amino Acids, Sulfur/metabolism , Amino Acids, Sulfur/blood , Metabolome , Gene Expression Regulation
3.
Oxid Med Cell Longev ; 2021: 9221693, 2021.
Article in English | MEDLINE | ID: mdl-34557267

ABSTRACT

OBJECTIVE: Aminothiols (glutathione (GSH), cysteinylglycine (CG)) may play an important role in the pathogenesis of coronavirus disease 2019 (COVID-19), but the possible association of these indicators with the severity of COVID-19 has not yet been investigated. METHODS: The total content (t) and reduced forms (r) of aminothiols were determined in patients with COVID-19 (n = 59) on admission. Lung injury was characterized by computed tomography (CT) findings in accordance with the CT0-4 classification. RESULTS: Low tGSH level was associated with the risk of severe COVID-19 (tGSH ≤ 1.5 µM, mild vs. moderate/severe: risk ratio (RR) = 3.09, p = 0.007) and degree of lung damage (tGSH ≤ 1.8 µM, CT < 2 vs. CT ≥ 2: RR = 2.14, p = 0.0094). The rGSH level showed a negative association with D-dimer levels (ρ = -0.599, p = 0.014). Low rCG level was also associated with the risk of lung damage (rCG ≤ 1.3 µM, CT < 2 vs. CT ≥ 2: RR = 2.28, p = 0.001). Levels of rCG (ρ = -0.339, p = 0.012) and especially tCG (ρ = -0.551, p = 0.004) were negatively associated with platelet count. In addition, a significant relationship was found between the advanced oxidation protein product level and tGSH in patients with moderate or severe but not in patients with mild COVID-19. CONCLUSION: Thus, tGSH and rCG can be seen as potential markers for the risk of severe COVID-19. GSH appears to be an important factor to oxidative damage prevention as infection progresses. This suggests the potential clinical efficacy of correcting glutathione metabolism as an adjunct therapy for COVID-19.


Subject(s)
COVID-19/diagnosis , Dipeptides/blood , Glutathione/blood , Advanced Oxidation Protein Products/blood , Aged , Amino Acids, Sulfur/blood , Biomarkers/blood , COVID-19/blood , COVID-19/pathology , Dipeptides/metabolism , Female , Glutathione/metabolism , Humans , Lung/diagnostic imaging , Lung/pathology , Male , Middle Aged , Oxidation-Reduction , SARS-CoV-2/isolation & purification , Severity of Illness Index
4.
J Nutr ; 151(4): 785-799, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33512502

ABSTRACT

BACKGROUND: Dietary sulfur amino acid restriction (SAAR) improves body composition and metabolic health across several model organisms in part through induction of the integrated stress response (ISR). OBJECTIVE: We investigate the hypothesis that activating transcription factor 4 (ATF4) acts as a converging point in the ISR during SAAR. METHODS: Using liver-specific or global gene ablation strategies, in both female and male mice, we address the role of ATF4 during dietary SAAR. RESULTS: We show that ATF4 is dispensable in the chronic induction of the hepatokine fibroblast growth factor 21 while being essential for the sustained production of endogenous hydrogen sulfide. We also affirm that biological sex, independent of ATF4 status, is a determinant of the response to dietary SAAR. CONCLUSIONS: Our results suggest that auxiliary components of the ISR, which are independent of ATF4, are critical for SAAR-mediated improvements in metabolic health in mice.


Subject(s)
Activating Transcription Factor 4/metabolism , Amino Acids, Sulfur/deficiency , Activating Transcription Factor 4/deficiency , Activating Transcription Factor 4/genetics , Amino Acids, Sulfur/blood , Amino Acids, Sulfur/metabolism , Animals , Antioxidants/metabolism , Body Composition , DNA/biosynthesis , Diet Therapy , Female , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/metabolism , Gene Knockdown Techniques , Hydrogen Sulfide/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Biosynthesis , Sex Factors , Stress, Physiological
5.
Stroke ; 52(1): 172-180, 2021 01.
Article in English | MEDLINE | ID: mdl-33349021

ABSTRACT

BACKGROUND AND PURPOSE: B-vitamin supplements lower circulating concentrations of homocysteine and may reduce stroke incidence. Homocysteine concentrations are associated with the incidence of stroke but other sulfur-containing compounds in the related metabolic pathway have not yet been investigated for an association with incident cerebrovascular diseases. METHODS: Nested within the EPIC (European Prospective Investigation Into Cancer and Nutrition)-Norfolk cohort, we established a case-control study with 480 incident cases of cerebrovascular diseases and 480 controls matched by age, sex, and year of baseline examination (1993-1997). Using baseline plasma samples, we assayed sulfur-containing compounds including methionine, homocysteine, cystathionine, cysteine, glutathione, and taurine with liquid chromatography-tandem mass spectrometry. We examined the association of concentrations of each of the compounds and the ratio of methionine to homocysteine (representing activity of one-carbon metabolism) with risk of incident cerebrovascular diseases, adjusted for potential confounders. RESULTS: Plasma methionine and the methionine/homocysteine ratio were inversely associated with risk of cerebrovascular diseases, with odds ratios per 1 SD of 0.83 (95% CI, 0.72-0.96) and 0.82 (95% CI, 0.71-0.95), respectively. The association of methionine remained significant after adjustment for homocysteine. None of the other examined compounds was significantly associated with incident cerebrovascular diseases. CONCLUSIONS: These findings suggest that greater availability of methionine, an essential amino acid, may play a role in the prevention of cerebrovascular diseases and explain the previously recognized link between elevated homocysteine and stroke. Further research is needed to determine causation and the potential of circulating methionine as a target in cerebrovascular disease prevention.


Subject(s)
Cerebrovascular Disorders/blood , Methionine/blood , Aged , Amino Acids, Sulfur/blood , Case-Control Studies , Cerebrovascular Disorders/epidemiology , Cohort Studies , Female , Humans , Incidence , Male , Middle Aged , Risk Factors
6.
J Nutr ; 150(Suppl 1): 2532S-2537S, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33000156

ABSTRACT

Homocysteine (Hcy) is methylated by methionine synthase to form methionine with methyl-cobalamin as a cofactor. The reaction demethylates 5-methyltetrahydrofolate to tetrahydrofolate, which is required for DNA and RNA synthesis. Deficiency of either of the cobalamin (Cbl) and/or folate cofactors results in elevated Hcy and megaloblastic anemia. Elevated Hcy is a sensitive biomarker of Cbl and/or folate status and more specific than serum vitamin assays. Elevated Hcy normalizes when the correct vitamin is given. Elevated Hcy is associated with alcohol use disorder and drugs that target folate or Cbl metabolism, and is a risk factor for thrombotic vascular disease. Elevated methionine and cystathionine are associated with liver disease. Elevated Hcy, cystathionine, and cysteine, but not methionine, are common in patients with chronic renal failure. Higher cysteine predicts obesity and future weight gain. Serum S-adenosylhomocysteine (AdoHcy) is elevated in Cbl deficiency and chronic renal failure. Drugs that require methylation for catabolism may deplete liver S-adenosylmethionine and raise AdoHcy and Hcy. Deficiency of Cbl or folate or perturbations of their metabolism cause major changes in sulfur amino acids.


Subject(s)
Amino Acids, Sulfur/metabolism , Folic Acid Deficiency/complications , Folic Acid/blood , Hyperhomocysteinemia/blood , Nutritional Status , Vitamin B 12 Deficiency/complications , Vitamin B 12/blood , Alcoholism/blood , Amino Acids, Sulfur/blood , Anemia, Megaloblastic/blood , Biomarkers/blood , Cardiovascular Diseases/blood , Folic Acid Deficiency/blood , Humans , Hyperhomocysteinemia/complications , Kidney Failure, Chronic/blood , Liver Diseases/blood , Obesity/blood , S-Adenosylhomocysteine/blood , Vitamin B 12 Deficiency/blood
7.
Biochimie ; 173: 91-99, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32330571

ABSTRACT

Homozygosity for the C677T polymorphism in MTHFR (TT genotype) is associated with a 24-87% increased risk of hypertension. Blood pressure (BP) lowering was previously reported in adults with the TT genotype, in response to supplementation with the MTHFR cofactor, riboflavin. Whether the BP phenotype associated with the polymorphism is related to perturbed one-carbon metabolism is unknown. This study investigated one-carbon metabolites and their responsiveness to riboflavin in adults with the TT genotype. Plasma samples from adults (n 115) screened for the MTHFR genotype, who previously participated in RCTs to lower BP, were analysed for methionine, S-adenosylmethionine (SAM), S-adenosylhomocysteine (SAH), betaine, choline and cystathionine by liquid chromatography tandem mass spectrometry (LC-MS/MS). The one-carbon metabolite response to riboflavin (1.6 mg/d; n 24) or placebo (n 23) for 16 weeks in adults with the TT genotype was also investigated. Plasma SAM (74.7 ± 21.0 vs 85.2 ± 22.6 nmol/L, P = 0.013) and SAM:SAH ratio (1.66 ± 0.55 vs 1.85 ± 0.51, P = 0.043) were lower and plasma homocysteine was higher (P = 0.043) in TT, compared to CC individuals. In response to riboflavin, SAM (P = 0.008) and cystathionine (P = 0.045) concentrations increased, with no responses in other one-carbon metabolites observed. These findings confirm perturbed one-carbon metabolism in individuals with the MTHFR 677TT genotype, and for the first time demonstrate that SAM, and cystathionine, increase in response to riboflavin supplementation in this genotype group. The genotype-specific, one-carbon metabolite responses to riboflavin intervention observed could offer some insight into the role of this gene-nutrient interaction in blood pressure.


Subject(s)
Amino Acids, Sulfur/blood , Betaine/blood , Choline/blood , Hypertension/blood , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Riboflavin/administration & dosage , Dietary Supplements , Female , Humans , Hypertension/metabolism , Male , Middle Aged , Mutation , Polymorphism, Genetic
8.
Biochimie ; 173: 107-113, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32147513

ABSTRACT

Evidence from human, animal and cellular studies suggests that high plasma total cysteine (tCys) is causally linked to human obesity, but determinants of population tCys variability are unknown. We hypothesized that tCys elevation in obesity may be mediated by an altered tCys response to intake of its precursor, methionine. We investigated whether BMI influences the change in plasma tCys, total homocysteine (tHcy) and total cysteinylglycine (tCysGly) 6h following a 100 mg/kg oral methionine load in 800 healthy subjects and 750 cardiovascular disease (CVD) cases. Methionine loading decreased tCys from mean 275 (95% CI, 273, 277) µmol/L to 253 (251,255) µmol/L. The decline in tCys was less in overweight (-8%) and obese (-6%) compared to normal weight (-9%) subjects, adjusting for age, gender and CVD (P-ANOVA = 0.006). Compared to normal weight subjects, individuals with obesity had a 2.8-fold likelihood (95% CI, 1.52, 5.01) of experiencing a rise (rather than decline), in tCys postload, after multiple adjustments. tCysGly also decreased postload, and the decline was similarly smaller in overweight (-18%) and obese (-15%) compared to normal weight (-21%) individuals (P-ANOVA <0.001). The tHcy response was modified by CVD status, with an increase in tHcy postload being BMI-dependent in controls (P-ANOVA<0.001) but not in CVD cases (P-interaction = 0.07). Although the methionine dose used was supraphysiologic, these data suggest that an altered tCys response to ingested methionine occurs in obesity, whereby tCys might rise in response to dietary methionine. This may contribute to explaining why human obesity is consistently associated with elevated tCys.


Subject(s)
Amino Acids, Sulfur/blood , Body Mass Index , Cardiovascular Diseases/blood , Methionine/administration & dosage , Adult , Case-Control Studies , Female , Humans , Male , Middle Aged , Obesity/blood , Overweight/blood
9.
Methods Mol Biol ; 2007: 9-17, 2019.
Article in English | MEDLINE | ID: mdl-31148103

ABSTRACT

In the context of the vascular effects of hydrogen sulfide (H2S), it is known that this gaseous endogenous biological modulator of inflammation, oxidative stress, etc. is a potent vasodilator. Chronic renal failure, a common disease affecting the aging population, is characterized by low levels of H2S in plasma and tissues, which could mediate their typical hypertensive pattern, along with other abnormalities. Lanthionine and homolanthionine, natural non-proteinogenic amino acids, are formed as side products of H2S production. Also in consideration of the intrinsic difficulties in H2S measuring, these compounds have been proposed as reliable and stable markers of H2S synthesis. However, in the setting of chronic renal failure patients on hemodialysis, they represent typical retention products (without ruling out the possibility of an increased intestinal synthesis) and prospective novel uremic toxins. Here, a method utilizing liquid chromatography-electrospray tandem mass spectrometry (LC-MS/MS) in multiple reaction monitoring ion mode has been developed and evaluated for the determination of these key H2S metabolites in plasma, by using a triple quadrupole mass spectrometer.


Subject(s)
Alanine/analogs & derivatives , Amino Acids, Sulfur/blood , Hydrogen Sulfide/blood , Renal Insufficiency, Chronic/blood , Sulfides/blood , Tandem Mass Spectrometry/methods , Alanine/blood , Chromatography, Liquid/methods , Humans , Male , Renal Dialysis , Renal Insufficiency, Chronic/therapy
10.
Nutrients ; 11(1)2018 Dec 20.
Article in English | MEDLINE | ID: mdl-30577516

ABSTRACT

Plasma sulphur-containing amino acids and related metabolites are associated with insulin sensitivity, although the mechanisms are unclear. We examined the effect of exercise on this relationship. Dysglycemic (n = 13) and normoglycemic (n = 13) men underwent 45 min cycling before and after 12 weeks exercise intervention. We performed hyperinsulinemic euglycemic clamp, mRNA-sequencing of skeletal muscle and adipose tissue biopsies, and targeted profiling of plasma metabolites by LC-MS/MS. Insulin sensitivity increased similarly in dysglycemic and normoglycemic men after 12 weeks of exercise, in parallel to similar increases in concentration of plasma glutamine, and decreased concentrations of plasma glutamate, cysteine, taurine, and glutathione. Change in plasma concentrations of cysteine and glutathione exhibited the strongest correlations to exercise-improved insulin sensitivity, and expression of a cluster of genes essential for oxidative phosphorylation and fatty acid metabolism in both skeletal muscle and adipose tissue, as well as mitochondria-related genes such as mitofilin. Forty-five min of cycling decreased plasma concentrations of glutamine and methionine, and increased plasma concentrations of glutamate, homocysteine, cystathionine, cysteine, glutathione, and taurine. Similar acute responses were seen in both groups before and after the 12 weeks training period. Both acute and long-term exercise may influence transsulphuration and glutathione biosynthesis, linking exercise-improved insulin sensitivity to oxidative stress and mitochondrial function.


Subject(s)
Amino Acids, Sulfur/blood , Exercise/physiology , Insulin Resistance/physiology , Overweight/blood , Adipose Tissue/metabolism , Bicycling , Exercise Test , Glucose Clamp Technique , Glutathione/metabolism , Humans , Insulin/blood , Male , Middle Aged , Muscle, Skeletal/metabolism , Overweight/physiopathology , RNA, Messenger/metabolism
11.
Nutrients ; 10(12)2018 Nov 23.
Article in English | MEDLINE | ID: mdl-30477080

ABSTRACT

Dietary and plasma total cysteine (tCys) have been associated with adiposity, possibly through interaction with stearoyl⁻CoA desaturase (SCD), which is an enzyme that is involved in fatty acid and energy metabolism. We evaluated the effect of a dietary intervention with low cysteine and methionine and high polyunsaturated fatty acids (PUFAs) on plasma and urinary sulfur amino acids and SCD activity indices. Fourteen normal-weight healthy subjects were randomized to a seven-day diet low in cysteine and methionine and high in PUFAs (Cys/Metlow + PUFA), or high in saturated fatty acids (SFA), cysteine, and methionine (Cys/Methigh + SFA). Compared with the Cys/Methigh + SFA group, plasma methionine and cystathionine decreased (p-values < 0.05), whereas cystine tended to increase (p = 0.06) in the Cys/Metlow + PUFA group. Plasma total cysteine (tCys) was not significantly different between the groups. Urinary cysteine and taurine decreased in the Cys/Metlow + PUFA group compared with the Cys/Methigh + SFA group (p-values < 0.05). Plasma SCD-activity indices were not different between the groups, but the change in cystine correlated with the SCD-16 index in the Cys/Metlow + PUFA group. A diet low in methionine and cysteine decreased plasma methionine and urinary cysteine and taurine. Plasma tCys was unchanged, suggesting that compensatory mechanisms are activated during methionine and cysteine restriction to maintain plasma tCys.


Subject(s)
Cysteine/administration & dosage , Diet , Energy Metabolism , Fatty Acids, Unsaturated/administration & dosage , Methionine/administration & dosage , Stearoyl-CoA Desaturase/metabolism , Adiposity , Adult , Amino Acids, Sulfur/administration & dosage , Amino Acids, Sulfur/blood , Amino Acids, Sulfur/metabolism , Cystathionine/metabolism , Cysteine/blood , Cysteine/metabolism , Cystine/metabolism , Female , Humans , Male , Methionine/blood , Methionine/metabolism , Obesity/metabolism , Obesity/prevention & control , Pilot Projects , Taurine/metabolism , Young Adult
12.
Rapid Commun Mass Spectrom ; 32(22): 1941-1948, 2018 Nov 30.
Article in English | MEDLINE | ID: mdl-30117207

ABSTRACT

RATIONALE: Preclinical studies in the search for treatments for several neurodegenerative diseases have identified lanthionine ketimine (LK) and its monoethyl ester derivative (LKE) as potential candidates. An ultrahigh-pressure liquid chromatography/tandem mass spectrometry (UHPLC/MS/MS) assay was developed to evaluate bioavailability by measuring these compounds in mouse serum, whole blood and brain tissue. METHODS: Following administration of LKE to mice for 3 days in chow at 300 ppm, the animals were sacrificed, and LKE was extracted from serum, whole blood and brain tissues through protein precipitation using cold methanol. To enhance chromatographic separation and electrospray ionization, LK was methylated using diazomethane. Separations were carried out using C18 reversed-phase UHPLC, and quantitative measurements were obtained using on-line triple-quadruple mass spectrometry with positive ion electrospray ionization, collision-induced dissociation and selected reaction monitoring. Tolbutamide was used as internal standard. RESULTS: LKE showed good recovery ranging from 77-90% in serum and 82-88% in brain tissue. An eight-point standard curve ranging from 0.005 to 4.6 µM was linear (R2 0.998). The average LKE detected in mouse serum was 277.42 nM, while the concentration in whole blood was 38 nM. Neither LK nor LKE was detected in brain tissues. CONCLUSIONS: A rapid quantitative method to measure LKE in mouse serum, whole blood and brain tissues using UHPLC/MS/MS was developed and validated following FDA guidelines. This method is suitable for bioavailability and pharmacokinetic studies.


Subject(s)
Amino Acids, Sulfur/blood , Amino Acids, Sulfur/pharmacokinetics , Brain/metabolism , Tandem Mass Spectrometry/methods , Animals , Biological Availability , Chromatography, High Pressure Liquid/methods , Esters/blood , Esters/pharmacokinetics , Limit of Detection , Mice
13.
Mol Ther ; 26(3): 834-844, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29398487

ABSTRACT

Classical homocystinuria (HCU) is the most common inherited disorder of sulfur amino acid metabolism caused by deficiency in cystathionine beta-synthase (CBS) activity and characterized by severe elevation of homocysteine in blood and tissues. Treatment with dietary methionine restriction is not optimal, and poor compliance leads to serious complications. We developed an enzyme replacement therapy (ERT) and studied its efficacy in a severe form of HCU in mouse (the I278T model). Treatment was initiated before or after the onset of clinical symptoms in an effort to prevent or reverse the phenotype. ERT substantially reduced and sustained plasma homocysteine concentration at around 100 µM and normalized plasma cysteine for up to 9 months of treatment. Biochemical balance was also restored in the liver, kidney, and brain. Furthermore, ERT corrected liver glucose and lipid metabolism. The treatment prevented or reversed facial alopecia, fragile and lean phenotype, and low bone mass. In addition, structurally defective ciliary zonules in the eyes of I278T mice contained low density and/or broken fibers, while administration of ERT from birth partially rescued the ocular phenotype. In conclusion, ERT maintained an improved metabolic pattern and ameliorated many of the clinical complications in the I278T mouse model of HCU.


Subject(s)
Cystathionine beta-Synthase/administration & dosage , Enzyme Replacement Therapy , Homocystinuria/diagnosis , Homocystinuria/therapy , Phenotype , Amino Acids, Sulfur/blood , Amino Acids, Sulfur/metabolism , Animals , Cystathionine beta-Synthase/chemistry , Disease Models, Animal , Drug Evaluation, Preclinical , Glucose/metabolism , Homocystinuria/metabolism , Lipid Metabolism , Liver/drug effects , Liver/metabolism , Mice , Oxidative Stress , Polyethylene Glycols/chemistry
14.
Anim Sci J ; 89(2): 448-455, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29154397

ABSTRACT

Exposure to high ambient temperature is detrimental to the poultry industry. To understand the influence from a metabolic perspective, we investigated the effects of exposure to high ambient temperature on plasma low-molecular-weight metabolite levels in chicks using gas chromatography/mass spectrometry-based non-targeted metabolomic analysis. Heat exposure for 4 days suppressed growth and food intake. Of the 92 metabolites identified, the levels of 29 decreased, whereas the levels of nine increased. We performed an enrichment analysis on the identified metabolites and found 35 candidates for metabolic processes affected by heat exposure. Among them, the sulfur amino acid metabolic pathway was clearly detected and the levels of the following related metabolites were decreased: cystathionine, cysteine, cystine, homocysteine and hypotaurine. Changes in the kynurenine pathway in tryptophan metabolism, which is linked to the immune system and oxidative stress, were also observed: kynurenine and quinolinic acid levels increased, whereas nicotinamide levels decreased. These results suggest the possible involvement of various metabolic processes in heat-exposed chicks. Some of these metabolites would be important to understand the mechanism of biological responses to high ambient temperature in chicks.


Subject(s)
Amino Acids, Sulfur/metabolism , Chickens/blood , Chickens/metabolism , Environmental Exposure/adverse effects , Hot Temperature/adverse effects , Amino Acids, Sulfur/blood , Animals , Cystathionine/blood , Cystathionine/metabolism , Cysteine/blood , Cysteine/metabolism , Cystine/blood , Cystine/metabolism , Gas Chromatography-Mass Spectrometry , Homocysteine/blood , Homocysteine/metabolism , Kynurenine/blood , Kynurenine/metabolism , Molecular Weight , Niacinamide/blood , Niacinamide/metabolism , Oxidative Stress , Quinolinic Acid/blood , Quinolinic Acid/metabolism , Taurine/analogs & derivatives , Taurine/blood , Taurine/metabolism , Tryptophan/blood , Tryptophan/metabolism
15.
Anim Sci J ; 88(12): 2016-2024, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28776913

ABSTRACT

A study was carried out to investigate the effects of dietary methionine source and level on plasma free amino acids patterns and the expression of genes involved in hepatic methionine metabolism in broiler breeders. A total of 2184 broiler breeders were assigned to 13 dietary treatments, with eight replicates per treatment. The 13 treatments included one control group and 12 additional treatments employing two sources and six levels (0.05, 0.10, 0.15, 0.20, 0.25 and 1.00%). Higher plasma methionine concentration was measured for DL-methionine (DLM) treated hens. Plasma alanine concentration was linearly increased as DLM or 2-hydroxy-4-(methylthio) butanoic acid (HMTBA) supplementation level increased. There was a linear increase in concentrations of tyrosine, valine, glycine and serine as dietary DLM supplementation level increased. Hens treated with DLM had higher relative expression of ADA than those fed HMTBA. The expression of MS, ADA, SAHH and MAT2A changed quadratically as HMTBA supplementation level increased, while the expression of GNMT and SAHH changed quadratically as DLM supplementation level increased. In conclusion, the effects of HMTBA on plasma free amino acid patterns and the expression of hepatic genes involved with methionine are different from DLM.


Subject(s)
Amino Acids, Sulfur/metabolism , Chickens/blood , Chickens/metabolism , Diet/veterinary , Dietary Supplements , Gene Expression , Liver/metabolism , Methionine/administration & dosage , Methionine/metabolism , Adenosine Deaminase , Alanine/blood , Amino Acids, Sulfur/blood , Animals , Butyrates/administration & dosage , Female , Glycine/blood , Methionine/blood , Methionine Adenosyltransferase , Serine/blood , Tyrosine/blood , Valine/blood
16.
Clin Chim Acta ; 473: 82-88, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28801090

ABSTRACT

BACKGROUND: We describe body composition, lipid metabolism and Stearoyl-CoA desaturase-1 (SCD-1) indices in patients with classical homocystinuria (HCU). METHODS: Eleven treated HCU patients and 16 healthy controls were included. Body composition and bone mineral density were assessed by dual X-ray absorptiometry. Sulfur amino acids (SAA) and their derivatives (total homocysteine, cysteine, methionine, S-adenosylmethionine, S-adenosylhomocysteine, and glutathione), lipids (free fatty acids, acylcarnitines, triglycerides and lipoproteins), glucose, insulin, leptin, adiponectin, and isoprostanes were measured in plasma. Insulin resistance was evaluated by HOMA-IR. To estimate liver SCD-1 activity, SCD-16 [16:1(n-7)/16:0] and SCD-18 [18:1(n-9)/18:0] desaturation indices were determined. RESULTS: In HCU patients, SCD-16 index was significantly reduced (p=0.03). A trend of an association of SCD-16 index with cysteine was observed (r=0.624, p=0.054). HCU patients displayed lower lean mass (p<0.05), with no differences in fat mass percentage. Leptin and low-density lipoprotein concentrations were lower in HCU patients (p<0.05). Femur bone mineral density Z-scores were correlated with plasma cysteine (r=0.829; p=0.04) and total homocysteine (r=-0.829; p=0.04) in HCU patients. CONCLUSIONS: We report alterations in leptin and SCD-1 in HCU patients. These results agree with previous findings from epidemiologic and animal studies, and support a role for SAA on lipid homeostasis.


Subject(s)
Amino Acids, Sulfur/blood , Homocystinuria/blood , Leptin/blood , Lipid Metabolism , Stearoyl-CoA Desaturase/blood , Adult , Bone Density , Female , Homocystinuria/metabolism , Homocystinuria/physiopathology , Humans , Male , Young Adult
17.
JAMA Psychiatry ; 73(6): 606-13, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27120188

ABSTRACT

IMPORTANCE: Vitamin B12, folate, and sulfur amino acids may be modifiable risk factors for structural brain changes that precede clinical dementia. OBJECTIVE: To investigate the association of circulating levels of vitamin B12, red blood cell folate, and sulfur amino acids with the rate of total brain volume loss and the change in white matter hyperintensity volume as measured by fluid-attenuated inversion recovery in older adults. DESIGN, SETTING, AND PARTICIPANTS: The magnetic resonance imaging subsample of the Swedish National Study on Aging and Care in Kungsholmen, a population-based longitudinal study in Stockholm, Sweden, was conducted in 501 participants aged 60 years or older who were free of dementia at baseline. A total of 299 participants underwent repeated structural brain magnetic resonance imaging scans from September 17, 2001, to December 17, 2009. MAIN OUTCOMES AND MEASURES: The rate of brain tissue volume loss and the progression of total white matter hyperintensity volume. RESULTS: In the multi-adjusted linear mixed models, among 501 participants (300 women [59.9%]; mean [SD] age, 70.9 [9.1] years), higher baseline vitamin B12 and holotranscobalamin levels were associated with a decreased rate of total brain volume loss during the study period: for each increase of 1 SD, ß (SE) was 0.048 (0.013) for vitamin B12 (P < .001) and 0.040 (0.013) for holotranscobalamin (P = .002). Increased total homocysteine levels were associated with faster rates of total brain volume loss in the whole sample (ß [SE] per 1-SD increase, -0.035 [0.015]; P = .02) and with the progression of white matter hyperintensity among participants with systolic blood pressure greater than 140 mm Hg (ß [SE] per 1-SD increase, 0.000019 [0.00001]; P = .047). No longitudinal associations were found for red blood cell folate and other sulfur amino acids. CONCLUSIONS AND RELEVANCE: This study suggests that both vitamin B12 and total homocysteine concentrations may be related to accelerated aging of the brain. Randomized clinical trials are needed to determine the importance of vitamin B12 supplementation on slowing brain aging in older adults.


Subject(s)
Alzheimer Disease/diagnostic imaging , Alzheimer Disease/physiopathology , Amino Acids, Sulfur/blood , Brain/diagnostic imaging , Folic Acid/blood , Magnetic Resonance Imaging , Vitamin B 12/blood , Aged , Atrophy , Brain/pathology , Female , Homocysteine/blood , Humans , Linear Models , Longitudinal Studies , Male , Middle Aged , Risk Factors , Statistics as Topic , Sweden , White Matter/diagnostic imaging
18.
Ann N Y Acad Sci ; 1363: 18-25, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26647293

ABSTRACT

The homocysteine theory of arteriosclerosis received credence when it was shown that after a methionine load, circulating homocysteine-cysteine concentrations were higher in cardiovascular disease patients than in healthy controls. Subsequent studies showing associations between homocysteine and coronary artery disease, stroke and cognitive impairment, relied on small increases in homocysteine concentration unlike the very high homocysteine seen in the rare genetic disorders that lead to homocystinuria and much higher homocysteine levels. Subsequent studies in cell culture, animals, and humans showed that a variety of cardiovascular adverse effects of "high homocysteine" introduced either as a nonphysiological bolus or as a methionine load led to high homocysteine. We fed apolipoprotein E-deficient mice diets designed to achieve three conditions: (1) high methionine intake with normal blood homocysteine, (2) high methionine intake with B vitamin deficiency and hyperhomocysteinemia, and (3) normal methionine intake with both B vitamin deficiency and hyperhomocysteinemia. We found that the mice fed methionine-rich diets had significant atheromatous pathology in the aortic arch even with normal plasma homocysteine levels. Mice fed B vitamin-deficient diets developed severe hyperhomocysteinemia but without any increase in vascular pathology. Our findings suggest that even moderate increases in methionine intake are atherogenic in susceptible mice while high plasma homocysteine is not.


Subject(s)
Amino Acids, Sulfur/metabolism , Atherosclerosis/etiology , Atherosclerosis/metabolism , Amino Acids, Sulfur/blood , Animals , Apolipoproteins E/deficiency , Atherosclerosis/blood , Atherosclerosis/pathology , Biomarkers , Blood Chemical Analysis , Diet , Disease Models, Animal , Homocysteine/blood , Homocysteine/metabolism , Metabolic Networks and Pathways , Methionine/blood , Methionine/metabolism , Mice , Mice, Knockout , Plaque, Atherosclerotic/pathology , Time Factors
19.
Eur J Nutr ; 55(1): 117-26, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25573689

ABSTRACT

PURPOSE: Although amino acids (AA) are required for growth, little is known about the effect of a deficient AA supply on the composition and the contractile and metabolic properties of skeletal muscles. METHODS: Protein metabolism, oxidative catabolism, glutathione system, and fiber-type composition of the longissimus (LM), rhomboideus (RM), and semitendinous (SM) muscles were compared between 42-day-old piglets pair-fed for 10 days either with a diet with a 28% deficient supply of total sulfur AA (TSAA-) or with a diet with a sufficient supply of total sulfur AA (TSAA+). RESULTS: The relative weight, protein mass, and protein synthesis of LM were 10-32% lower in TSAA- pigs compared with TSAA+ pigs, while RM and SM were not affected by the TSAA supply. The TSAA supply affected the AA composition of muscles. Concentrations of Met and branched-chain AA were, respectively, 7 and 3% lower in TSAA- pigs compared with TSAA+ pigs. The His concentration was 30% higher in LM and SM in TSAA- pigs compared with TSAA+ pigs and unaffected in RM. The activity of citrate synthase was 14% higher in all three muscles of TSAA- pigs. In these pigs, the ß-hydroxy-acyl-CoA dehydrogenase activity was 20% higher in RM compared with TSAA+ pigs while that of lactate dehydrogenase was 21% lower in LM. Total and reduced glutathione concentrations were more than 70% greater in RM than in LM or SM, and these concentrations were approximately 10% lower in TSAA- pigs than in TSAA+ pigs. CONCLUSIONS: Results of this study indicate that a TSAA deficiency affects muscle properties in a muscle-dependent manner increasing the oxidative capacity of RM and reducing growth and glycolytic metabolism of LM.


Subject(s)
Amino Acids, Sulfur/administration & dosage , Animal Feed/analysis , Diet/veterinary , Muscle, Skeletal/metabolism , Amino Acids, Sulfur/blood , Amino Acids, Sulfur/deficiency , Animals , Body Weight , Energy Metabolism , Glutathione/metabolism , L-Lactate Dehydrogenase/metabolism , Muscle, Skeletal/drug effects , Protein Biosynthesis , Sus scrofa/growth & development
20.
São Paulo; s.n; 2015. 117 p.
Thesis in Portuguese | LILACS | ID: lil-781882

ABSTRACT

Em diversos países, inclusive no Brasil, a fortificação de alimentos com ácido fólico (AF) foi adotada como política pública de prevenção e combate à deficiência nutricional da vitamina, motivados principalmente pela redução da incidência dos defeitos do tubo neural. No período pós-fortificação observa-se tanto a evolução positiva do consumo e nível sérico da vitamina quanto a diminuição da concentração plasmática de homocisteína, e ainda, o aumento do ácido fólico não metabolizado na maioria desses países. Não se conhece ainda os efeitos biológicos do AFNM, no entanto, considera-se que o AFNM pode ser um fator relevante nas questões de segurança associadas com alto consumo de AF. Objetivo: Avaliar o consumo dietético e nível de folato, homocisteína e ácido fólico não metabolizado após a fortificação mandatória de alimentos com ácido fólico, e a interação com os polimorfismos envolvidos no metabolismo do folato e homocisteína. Metodologia: Os dados foram oriundos do estudo transversal de base populacional ISA Capital 2008 conduzido em uma amostra representativa de residentes do município de São Paulo, de ambos os sexos, e com idade acima de 14 anos. Coletou-se recordatórios alimentares de 24 horas e amostra de sangue em jejum de 12 horas para análises bioquímicas e moleculares. As análises estatísticas foram realizadas no programa STATA®, versão 13.0, com nível de significância de 5 por cento . Resultados: O estudo foi conduzido em 750 indivíduos, sendo 53,1 por cento mulheres e média de idade 40,7 (IC95 por cento 38,8-42,5) anos. A média de consumo e nível de folato foi de 375,8 (IC95 por cento 363,0-388,6) mcg/dia e 13 (IC95 por cento 12,0-13,9) ng/ml, respectivamente...


Food fortification is an important strategy in public health policy for controlling micronutrient malnutrition, and a major contributory factor in the eradication of micronutrients deficiencies. Motivated by the reduction in the occurrence of neural tube defects, countries worldwide, including Brazil, adopted food fortification with folic acid (FA). Folic acid fortification has increased dietary intakes of folic acid and folate status, but it is also associated with the presence of circulating FA. Although the metabolism and biological effects of circulating of folic acid are not well known, it may be a contributing factor in safety concerns associated with high oral doses of folic acid. Objective: To assess the folate intake and status, homocysteine and circulating FA after mandatory fortification with folic acid, and interaction with polymorphisms involved in 1-carbon metabolism. Material and Methods: Data were from 750 individuals aged > 14 years old who participated of a cross-sectional population-based survey in Sao Paulo City-Brazil. Fasting blood samples and information about food intake based on two measures of 24 hour food recall were collected. All analyses were carried out using STATA (version 13.0) and p-value <.05 was considered to be statistically significant in all tests. Results: Results were from 750 individuals...


Subject(s)
Humans , Male , Female , Adolescent , Adult , Middle Aged , Folic Acid/metabolism , Amino Acids, Sulfur/metabolism , Eating , Food, Fortified , Homocysteine/metabolism , Polymorphism, Genetic , Folic Acid/blood , Amino Acids, Sulfur/blood , Cross-Sectional Studies , Homocysteine/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...