Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Expert Rev Gastroenterol Hepatol ; 16(9): 879-894, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35929959

ABSTRACT

INTRODUCTION: Homocysteine is a sulfur-containing amino acid formed in the intermediary metabolism of methionine. Amino acid metabolism and heme biosynthesis pathways are complexly intertwined. Plasma homocysteine elevation, hyperhomocysteinemia (HHcy), has been reported in patients with acute hepatic porphyria (AHP), a family of rare genetic disorders caused by defects in hepatic heme biosynthesis. AREAS COVERED: This article summarizes published case series in which givosiran, a subcutaneously administered small interfering RNA approved for AHP treatment, appeared to exacerbate dysregulated homocysteine metabolism in patients with AHP. A comprehensive exploratory analysis of ENVISION trial data demonstrated that on a population level, givosiran increased homocysteine but with wide interpatient variations, and there is no proof of correlations between HHcy and changes in efficacy or safety of givosiran. EXPERT OPINION: The strong correlation and co-increase of homocysteine and methionine suggest that HHcy associated with givosiran is likely attributable to the impaired trans-sulfuration pathway catalyzed by cystathionine ß-synthase, which uses vitamin B6 as a cofactor. Data-based consensus supports monitoring total plasma homocysteine and vitamin B6, B12, and folate levels before and during givosiran treatment; supplementing with pyridoxine/vitamin B6 in patients with homocysteine levels >100 µmol/L; and involving patients with homocysteine levels >30 µmol/L in decisions to supplement.


Subject(s)
Hyperhomocysteinemia , Porphyrias, Hepatic , Humans , Cystathionine beta-Synthase/genetics , Folic Acid , Heme , Homocysteine , Hyperhomocysteinemia/diagnosis , Hyperhomocysteinemia/drug therapy , Methionine/metabolism , Porphyrias, Hepatic/diagnosis , Porphyrias, Hepatic/drug therapy , Porphyrias, Hepatic/complications , Pyridoxine , RNA, Small Interfering , Sulfur , Vitamin B 6 , Clinical Trials as Topic
2.
Zhonghua Er Ke Za Zhi ; 60(6): 533-538, 2022 Jun 02.
Article in Chinese | MEDLINE | ID: mdl-35658358

ABSTRACT

Objective: To analyze the clinical features and CBS gene variants of 13 patients with classic homocystinuria, and the strategies of individual treatment and prevention were explored. Methods: The general information, clinical manifestations, laboratory tests, cranial images, CBS gene variants, diagnosis and therapeutic strategies of 13 patients with classic homocystinuria admitted to the Department of Pediatrics of Children's Hospital Affiliated to Zhengzhou University and Peking University First Hospital from November 2013 to June 2021 were analyzed retrospectively. Results: There were 13 patients diagnosed at the age of 10 days to 14 years, 6 were male and 7 were female. There were 3 patients detected by newborn screening and received treatment at the asymptomatic stage. There were 10 patients clinically diagnosed at the age of 5 to 14 years. Their symptoms appeared at age of 1 to 6 years. The major clinical manifestations were marfanoid features, lens dislocation and (or) myopia, developmental delay, osteoporosis, and cardiovascular diseases. Brain magnetic resonance imaging showed asymmetric infarcts in 4 patients and hypomyelination in 1 case. Increased blood methionine, plasma total homocysteine and urinary total homocysteine with normal urinary methylmalonic acid were found in 13 patients. The biochemical features were consistent with classic homocystinuria. Totally 18 variants were identified in CBS gene of 13 patients, 10 variants were novel and 8 were reported. only 1 patient was partially responsive to vitamin B6 treatment, while 12 cases were non-responsive. They were mainly treated with low methionine diet and betaine supplement. Three vitamin B6 non-responsive cases received liver transplantation at age of 3, 8 and 8 years, respectively. Their blood methionine and total homocysteine returned to normal within a week after liver transplantation. One patient died. Prenatal diagnosis was performed for a fetus when the mother was pregnant again. Two pathogenic CBS gene variants were identified from the amniocytes as same as the proband. Conclusions: The clinical manifestations of classic homocystinuria are complex and variable. Blood amino acid analysis, serum or urine total homocysteine assay and gene analysis are critical for its diagnosis. There were 10 novel CBS gene varients were identified expanding the CBS gene varient spectrum. Liver transplantation is an effective treatment. Prenatal diagnosis is important to prevent classic homocysteinuria.


Subject(s)
Homocystinuria , Adolescent , Child , Child, Preschool , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/therapeutic use , Female , Homocysteine/therapeutic use , Homocystinuria/diagnosis , Homocystinuria/drug therapy , Homocystinuria/genetics , Humans , Infant , Infant, Newborn , Male , Methionine/therapeutic use , Pyridoxine/therapeutic use , Retrospective Studies , Vitamins/therapeutic use
3.
J Inherit Metab Dis ; 44(6): 1382-1392, 2021 11.
Article in English | MEDLINE | ID: mdl-34528713

ABSTRACT

Cystathionine ß-synthase (CBS) deficiency is a recessive inborn error of sulfur metabolism characterized by elevated blood levels of total homocysteine (tHcy). Patients diagnosed with CBS deficiency are currently treated by a combination of vitamin supplementation and restriction of foods containing the homocysteine precursor methionine, but the effectiveness of this therapy is limited due to poor compliance. A mouse model for CBS deficiency (Tg-I278T Cbs-/- ) was used to evaluate a potential gene therapy approach to treat CBS deficiency utilizing an AAVrh.10-based vector containing the human CBS cDNA downstream of the constitutive, strong CAG promoter (AAVrh.10hCBS). Mice were administered a single dose of virus and followed for up to 1 year. The data demonstrated a dose-dependent increase in liver CBS activity and a dose-dependent decrease in serum tHcy. Liver CBS enzyme activity at 1 year was similar to Cbs+/- control mice. Mice given the highest dose (5.6 × 1011 genomes/mouse) had mean serum tHcy decrease of 97% 1 week after injection and an 81% reduction 1 year after injection. Treated mice had either full- or substantial correction of alopecia, bone loss, and fat mass phenotypes associated with Cbs deficiency in mice. Our findings show that AAVrh.10-based gene therapy is highly effective in treating CBS deficiency in mice and supports additional pre-clinical testing for eventual use human trials.


Subject(s)
Cystathionine beta-Synthase/genetics , Dependovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Homocystinuria/genetics , Homocystinuria/therapy , Animals , Cystathionine beta-Synthase/blood , Cystathionine beta-Synthase/deficiency , Disease Models, Animal , Female , Gene Expression , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Homocystinuria/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Phenotype
4.
Expert Rev Anticancer Ther ; 21(11): 1287-1298, 2021 11.
Article in English | MEDLINE | ID: mdl-34320879

ABSTRACT

Aim: To explore the clinical significance of Cystathionine beta-synthase (CBS) expression in gastric cancer (GC).Research design and methods: CBS expression and clinicopathological/follow-up information of patients with gastric cancer undergoing operation were collected from The Cancer Genome Atlas (TCGA) database. The association of CBS expression with patients' overall survival (OS) was determined in the entire cohort and different subgroups. Validation was performed in two external cohorts from NCBI Gene Expression Omnibus (GEO) database. The estimated drug response of the tumors with different CBS expressions was characterized. The potential CBS-related cellular pathways in chemoresistance were explored.Results: High CBS was associated with poor OS in patients receiving adjuvant chemotherapy (ACT) but not those without ACT. And ACT was associated with favorable OS in patients with low CBS expression but not those with high CBS expression. The results were verified in two external cohorts. Drug response prediction suggested that patients with low CBS expression showed high sensitivity to 5-Fluorouracil. Gene Set Enrichment Analysis (GSEA) suggested that CBS might contribute to GC chemoresistance via modulating many cellular pathways, including down-regulating apoptosis and P53 pathways while up-regulating DNA repair pathway.Conclusion: Low CBS expression can predict the benefit from ACT in GC.


Subject(s)
Cystathionine beta-Synthase , Stomach Neoplasms , Chemotherapy, Adjuvant , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/therapeutic use , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Prognosis , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
5.
Am J Med Genet A ; 185(4): 1247-1250, 2021 04.
Article in English | MEDLINE | ID: mdl-33443292

ABSTRACT

We report a patient with homocystinuria and hyperoxaluria who was cured of homocystinuria-related disease following liver transplant. The patient was diagnosed with homocystinuria as a newborn and was treated with dietary modifications and supplements. At 22 months, he passed a calcium oxalate stone and was found to have numerous bilateral kidney stones. Genetic testing confirmed primary hyperoxaluria, type 1. He underwent preemptive liver transplant at age four to treat primary hyperoxaluria. Following transplant, his serum methionine and homocysteine levels normalized, thus, demonstrating resolution of homocystinuria. Methionine and homocysteine levels remained normal 6 years later. Homocystinuria is associated with ophthalmologic, skeletal, neurologic, and thromboembolic complications. As cystathionine beta-synthase resides in the liver, transplant was hypothesized to be an effective treatment. Primary hyperoxaluria generally progresses to chronic kidney disease and is treated with combined kidney-liver transplant at the time of end stage kidney disease. Given this patient's dual diagnoses, we proceeded with preemptive liver transplantation. Three prior cases of patients with homocystinuria treated with liver transplantation have been reported. In all cases, transplant resolved metabolic effects. However, our case represents a pediatric patient without disease-related complications prior to transplant. This case supports liver-targeted gene therapies as an effective treatment for homocystinuria.


Subject(s)
Cystathionine beta-Synthase/genetics , Homocystinuria/genetics , Homocystinuria/therapy , Liver Transplantation , Cystathionine beta-Synthase/deficiency , Female , Homocysteine/blood , Homocystinuria/blood , Homocystinuria/pathology , Humans , Infant , Infant, Newborn , Male , Methionine/blood , Neonatal Screening , Pediatrics
6.
Sci Rep ; 10(1): 14657, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32887901

ABSTRACT

Cystathionine ß-synthase (CBS) catalyzes the condensation of serine and homocysteine to water and cystathionine, which is then hydrolyzed to cysteine, α-ketobutyrate and ammonia by cystathionine γ-lyase (CGL) in the reverse transsulfuration pathway. The protozoan parasite Toxoplasma gondii, the causative agent of toxoplasmosis, includes both CBS and CGL enzymes. We have recently reported that the putative T. gondii CGL gene encodes a functional enzyme. Herein, we cloned and biochemically characterized cDNA encoding CBS from T. gondii (TgCBS), which represents a first example of protozoan CBS that does not bind heme but possesses two C-terminal CBS domains. We demonstrated that TgCBS can use both serine and O-acetylserine to produce cystathionine, converting these substrates to an aminoacrylate intermediate as part of a PLP-catalyzed ß-replacement reaction. Besides a role in cysteine biosynthesis, TgCBS can also efficiently produce hydrogen sulfide, preferentially via condensation of cysteine and homocysteine. Unlike the human counterpart and similar to CBS enzymes from lower organisms, the TgCBS activity is not stimulated by S-adenosylmethionine. This study establishes the presence of an intact functional reverse transsulfuration pathway in T. gondii and demonstrates the crucial role of TgCBS in biogenesis of H2S.


Subject(s)
Cystathionine beta-Synthase/metabolism , Cysteine/biosynthesis , Hydrogen Sulfide/metabolism , Toxoplasma/enzymology , Toxoplasma/genetics , Biocatalysis , Cystathionine/biosynthesis , Cystathionine beta-Synthase/genetics , Cystathionine gamma-Lyase/metabolism , DNA, Complementary , Genes, Protozoan , Heme/metabolism , Homocysteine/metabolism , Kinetics , Serine/analogs & derivatives , Serine/metabolism
7.
Am J Med Genet A ; 182(11): 2704-2708, 2020 11.
Article in English | MEDLINE | ID: mdl-32820583

ABSTRACT

Classic homocystinuria is due to deficiency of cystathionine beta-synthase (CBS), a pyridoxine-dependent enzyme that, depending on the molecular variants, may be co-factor responsive. Elevated methionine is often used as the primary analyte to detect CBS deficiency (CBSD) on newborn screening (NBS), but is limited by increased detection of other biochemical disorders with less clear clinical significance such as methionine aminotransferase (MAT) I/III heterozygotes. Our state has implemented a two-tier NBS algorithm for CBSD that successfully reduced the number of MATI/III heterozygotes, yet effectively detected a mild, co-factor responsive form of CBSD. After initial diagnosis, newborns with CBSD often undergo a pyridoxine challenge with high-dose pyridoxine to determine responsiveness. Here we describe our NBS-identified patient with a mild form of pyridoxine responsive CBSD who developed respiratory failure and rhabdomyolysis consistent with pyridoxine toxicity during a pyridoxine challenge. This case highlights the need for weight-based dosing and duration recommendations for pyridoxine challenge in neonates.


Subject(s)
Cystathionine beta-Synthase/deficiency , Cystathionine beta-Synthase/genetics , Homocystinuria/drug therapy , Neonatal Screening/methods , Pyridoxine/adverse effects , Respiratory Insufficiency/pathology , Rhabdomyolysis/pathology , Dose-Response Relationship, Drug , Female , Homocystinuria/genetics , Homocystinuria/pathology , Humans , Infant, Newborn , Prognosis , Pyridoxine/administration & dosage , Respiratory Insufficiency/chemically induced , Rhabdomyolysis/chemically induced , Vitamin B Complex/administration & dosage , Vitamin B Complex/adverse effects
8.
Genes (Basel) ; 11(3)2020 03 20.
Article in English | MEDLINE | ID: mdl-32245022

ABSTRACT

Homocystinuria is an inborn error of metabolism due to the deficiency in cystathionine beta-synthase (CBS) enzyme activity. It leads to the elevation of both homocysteine and methionine levels in the blood and urine. Consequently, this build-up could lead to several complications such as nearsightedness, dislocated eye lenses, a variety of psychiatric and behavioral disorders, as well as vascular system complications. The prevalence of homocystinuria is around 1/200,000 births worldwide. However, its prevalence in the Gulf region, notably Qatar, is exceptionally high and reached 1:1800. To date, more than 191 pathogenic CBS mutations have been documented. The majority of these mutations were identified in Caucasians of European ancestry, whereas only a few mutations from African-Americans or Asians were reported. Approximately 87% of all CBS mutations are missense and do not target the CBS catalytic site, but rather result in unstable misfolded proteins lacking the normal biological function, designating them for degradation. The early detection of homocystinuria along with low protein and methionine-restricted diet is the best treatment approach for all types of homocystinuria patients. Yet, less than 50% of affected individuals show a significant reduction in plasma homocysteine levels after treatment. Patients who fail to lower the elevated homocysteine levels, through high protein-restricted diet or by B6 and folic acid supplements, are at higher risk for cardiovascular diseases, neurodegenerative diseases, neural tube defects, and other severe clinical complications. This review aims to examine the mutations spectrum of the CBS gene, the disease management, as well as the current and potential treatment approaches with a greater emphasis on studies reported in the Middle East and North Africa (MENA) region.


Subject(s)
Cystathionine beta-Synthase/genetics , Homocystinuria/genetics , Mutation , Genetic Testing/methods , Homocystinuria/diagnosis , Homocystinuria/epidemiology , Homocystinuria/therapy , Humans
9.
Acta Physiol (Oxf) ; 228(3): e13373, 2020 03.
Article in English | MEDLINE | ID: mdl-31483934

ABSTRACT

AIM: Whereas some patients have important changes in body core temperature (Tb) during systemic inflammation, others maintain a normal Tb, which is intrinsically associated to immune paralysis. One classical model to study immune paralysis is the use of repeated administration of lipopolysaccharide (LPS), the so-called endotoxin tolerance. However, the neuroimmune mechanisms of endotoxin tolerance remain poorly understood. Hydrogen sulphide (H2 S) is a gaseous neuromodulator produced in the brain by the enzyme cystathionine ß-synthase (CBS). The present study assessed whether endotoxin tolerance is modulated by hypothalamic H2 S. METHODS: Rats with central cannulas (drug microinjection) and intraperitoneal datalogger (temperature record) received a low-dose of lipopolysaccharide (LPS; 100 µg kg-1 ) daily for four consecutive days. Hypothalamic CBS expression and H2 S production rate were assessed, together with febrigenic signalling. Tolerant rats received an inhibitor of H2 S synthesis (AOA, 100 pmol 1 µL-1 icv) or its vehicle in the last day. RESULTS: Antero-ventral preoptic area of the hypothalamus (AVPO) H2 S production rate and CBS expression were increased in endotoxin-tolerant rats. Additionally, hypothalamic H2 S inhibition reversed endotoxin tolerance reestablishing fever, AVPO and plasma PGE2 levels without altering the absent plasma cytokines surges. CONCLUSION: Endotoxin tolerance is not simply a reflection of peripheral reduced cytokines release but actually results from a complex set of mechanisms acting at multiple levels. Hypothalamic H2 S production modulates most of these mechanisms.


Subject(s)
Dinoprostone/biosynthesis , Endotoxins/pharmacology , Hydrogen Sulfide/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Animals , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cytokines/metabolism , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Disease Models, Animal , Drug Tolerance , Fever/drug therapy , Fever/metabolism , Lipopolysaccharides/pharmacology , Male , Preoptic Area/drug effects , Preoptic Area/metabolism , Rats , Rats, Wistar
10.
Hum Gene Ther ; 30(9): 1093-1100, 2019 09.
Article in English | MEDLINE | ID: mdl-31084364

ABSTRACT

Cystathionine ß-synthase (CBS) deficiency is a recessive inborn error of metabolism characterized by extremely elevated total homocysteine (tHcy) in the blood. Patients diagnosed with CBS deficiency have a variety of clinical problems, including dislocated lenses, osteoporosis, cognitive and behavioral issues, and a significantly increased risk of thrombosis. Current treatment strategies involve a combination of vitamin supplementation and restriction of foods containing the homocysteine precursor methionine. Here, a mouse model for CBS deficiency (Tg-I278T Cbs-/-) was used to evaluate the potential of minicircle-based naked DNA gene therapy to treat CBS deficiency. A 2.3 kb DNA-minicircle containing the liver-specific P3 promoter driving the human CBS cDNA (MC.P3-hCBS) was delivered into Tg-I278T Cbs-/- mice via a single hydrodynamic tail vein injection. Mean serum tHcy decreased from 351 µM before injection to 176 µM 7 days after injection (p = 0.0005), and remained decreased for at least 42 days. Western blot analysis reveals significant minicircle-directed CBS expression in the liver tissue. Liver CBS activity increased 34-fold (12.8 vs. 432 units; p = 0.0004) in MC.P3-hCBS-injected animals. Injection of MC.P3-hCBS in young mice, subsequently followed for 202 days, showed that the vector can ameliorate the mouse homocystinuria alopecia phenotype. The present findings show that minicircle-based gene therapy can lower tHcy in a mouse model of CBS deficiency.


Subject(s)
Cystathionine beta-Synthase/genetics , DNA, Circular/genetics , Genetic Therapy , Genetic Vectors/genetics , Homocystinuria/genetics , Homocystinuria/therapy , Animals , Biomarkers , Cystathionine beta-Synthase/blood , Cystathionine beta-Synthase/deficiency , DNA, Circular/administration & dosage , Disease Models, Animal , Female , Gene Expression , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Homocystinuria/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Phenotype , Transfection/methods , Treatment Outcome
11.
J Ethnopharmacol ; 238: 111813, 2019 Jun 28.
Article in English | MEDLINE | ID: mdl-30910578

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Whitmania pigra Whitman (Whitmania pigra, WP), firstly recorded in the Shennong's Herbal Classic and officially listed in the Chinese Pharmacopoeia, is a well-used cardiovascular protective traditional Chinese medicine derived from leeches. Traditional Chinese physicians prefer to prescribe the dried whole body of leech processed under high temperatures. It has been reported that dried WP remains clinically effective. However, the therapeutic mechanism has yet not be clearly elucidated. AIM OF THE STUDY: This study was designed to investigate the protective activity of the extract of WP in a high-molecular-weight dextran-induced blood hyperviscosity rat model, and to explore the role of WP in improving blood hyperviscosity related metabolic disorders and to clarify the possible mechanism of metabolic regulation. MATERIALS AND METHODS: The hemorheological parameters were measured with an automated blood rheology analyzer. Hematoxylin-eosin staining was used to observe the pathological changes in aortic tissues samples. Further, a liquid chromatography-mass-spectrometry (LC-MS)-based untargeted metabolomics approach was applied to characterize the metabolic alterations. RESULTS: WP has evident attenuating effects on blood hyperviscosity and related metabolic disorders, and the influences are distinct from those of aspirin. The results showed that WP had good effects in reducing blood viscosity and ameliorating histopathological changes in the thoracic aorta in a high molecular weight dextran-induced blood hyperviscosity rat model. The middle dose (2.5 g raw material/kg body weight) of WP exhibited effects equivalent to aspirin (100 mg/kg) on hemorheological and histopathological parameters (P > 0.05). However, when using metabolomics profiling, we found that WP could significantly improve blood hyperviscosity-related metabolic disorders and restore metabolites to normal levels; while aspirin showed little effect. With principal component analysis and orthogonal partial least-squares discriminant analysis, WP regulated many more endogenous metabolites than aspirin. With pathway enrichment analysis, the differential endogenous metabolites were involved in cysteine and methionine metabolism, TCA cycle, arachidonic acid metabolism, etc., highlighting the metabolic reprogramming potential of WP against blood hyperviscosity-induced metabolic disorders. CONCLUSIONS: The study suggest that WP has a more potent effect, but a different mechanism, than aspirin in improving either blood hyperviscosity or related metabolic disorders associated with cardio- and cerebrovascular diseases.


Subject(s)
Blood Viscosity/drug effects , Complex Mixtures/pharmacology , Leeches , Animals , Cyclooxygenase 2/genetics , Cystathionine beta-Synthase/genetics , Liver/drug effects , Liver/metabolism , Male , Powders , Rats, Sprague-Dawley
12.
Mol Nutr Food Res ; 61(11)2017 11.
Article in English | MEDLINE | ID: mdl-28759161

ABSTRACT

SCOPE: Serine lies at the central node linking biosynthetic flux from glycolysis to glutathione synthesis and one-carbon metabolic cycle which are closely related to antioxidant capacity. The present study was conducted to determine the effects of serine supplementation on oxidative stress and its relative mechanisms. METHODS AND RESULTS: Diquat treatment was performed to induce oxidative stress in mice and primary hepatocytes. The results showed that hepatic glutathione anti-oxidant systems were impaired and reactive oxygen species and homocysteine were increased in diquat-induced mice and hepatocytes, while such disadvantageous changes were diminished by serine supplementation both in vivo and in vitro. However, when cystathionine ß-synthase expression was inhibited by interference RNA in hepatocytes, the effects of serine supplementation on the improvement of glutathione synthesis and the alleviation of oxidative stress were diminished. Moreover, when hepatocytes were treated with cycloleucine, an inhibitor of methionine adenosyltransferase, the effects of serine supplementation on the improvement of methionine cycle and the alleviation of DNA hypomethylation and oxidative stress were also diminished. CONCLUSION: Our results indicated that serine supplementation alleviated oxidative stress via supporting glutathione synthesis and methionine cycle, mostly by condensing with homocysteine to synthesize cysteine and providing one-carbon units for homocysteine remethylation.


Subject(s)
Antioxidants/therapeutic use , Dietary Supplements , Glutathione/metabolism , Hepatocytes/metabolism , Methionine/metabolism , Oxidative Stress , Serine/therapeutic use , Animals , Antioxidants/chemistry , Antioxidants/metabolism , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Cycloleucine/pharmacology , Cystathionine beta-Synthase/antagonists & inhibitors , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , DNA Methylation/drug effects , Defoliants, Chemical/antagonists & inhibitors , Defoliants, Chemical/toxicity , Diquat/antagonists & inhibitors , Diquat/toxicity , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/drug effects , Hepatocytes/pathology , Homocysteine/metabolism , Male , Methionine Adenosyltransferase/antagonists & inhibitors , Methionine Adenosyltransferase/metabolism , Mice, Inbred C57BL , Oxidative Stress/drug effects , RNA Interference , Random Allocation , Serine/antagonists & inhibitors , Serine/metabolism , Specific Pathogen-Free Organisms
13.
Oxid Med Cell Longev ; 2017: 8940321, 2017.
Article in English | MEDLINE | ID: mdl-28421128

ABSTRACT

The human disease classical homocystinuria results from mutations in the gene encoding the pyridoxal 5'-phosphate- (PLP-) dependent cystathionine ß-synthase (CBS), a key enzyme in the transsulfuration pathway that controls homocysteine levels, and is a major source of the signaling molecule hydrogen sulfide (H2S). CBS activity, contributing to cellular redox homeostasis, is positively regulated by S-adenosyl-L-methionine (AdoMet) but fully inhibited upon CO or NO• binding to a noncatalytic heme moiety. Despite extensive studies, the molecular basis of several pathogenic CBS mutations is not yet fully understood. Here we found that the ferrous heme of the reportedly mild p.P49L CBS variant has altered spectral properties and markedly increased affinity for CO, making the protein much more prone than wild type (WT) CBS to inactivation at physiological CO levels. The higher CO affinity could result from the slightly higher flexibility in the heme surroundings revealed by solving at 2.80-Å resolution the crystallographic structure of a truncated p.P49L. Additionally, we report that p.P49L displays impaired H2S-generating activity, fully rescued by PLP supplementation along the purification, despite a minor responsiveness to AdoMet. Altogether, the results highlight how increased propensity to CO inactivation of an otherwise WT-like variant may represent a novel pathogenic mechanism in classical homocystinuria.


Subject(s)
Cystathionine beta-Synthase/metabolism , Hydrogen Sulfide/metabolism , Carbon Monoxide/chemistry , Carbon Monoxide/metabolism , Crystallography, X-Ray , Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/genetics , Heme/chemistry , Heme/metabolism , Humans , Kinetics , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , S-Adenosylmethionine/metabolism
14.
Food Chem Toxicol ; 103: 183-187, 2017 May.
Article in English | MEDLINE | ID: mdl-28288933

ABSTRACT

Altered homocysteine metabolism defined as hyperhomocysteinemia is implicated as pathogenic factor in several cardiovascular diseases and atherosclerosis. The purpose of this study was to investigate the efficacy of prune extract, a good source of phenolic antioxidants, on lowering plasma homocysteine level in male hyperhomocysteinemic mice from average weight of 28 g. The administration of lyophilized prune extract was carried out by intraperitoneal injection one day preceding and one hour before sacrifice of mice. Prune extract decreased significantly plasma homocysteine level, correlated with an increased activity of S-adenosylhomocysteine (SAH) hydrolase and NAD(P)H: quinone oxydoreductase-1 activities. Our results suggest a beneficial effect of prune extract on hyperhomocysteinemia with reduction of homocysteine level by its conversion on to SAH by S-adenosylhomocysteine hydrolase, which is activated by NAD+, a by-product of NAD(P)H: quinone oxydo reductase-1.


Subject(s)
Hyperhomocysteinemia/diet therapy , Plant Extracts/pharmacology , Prunus domestica/chemistry , Adenosylhomocysteinase/metabolism , Animals , Chlorogenic Acid/pharmacology , Cystathionine beta-Synthase/genetics , Female , Freeze Drying , Homocysteine/blood , Hyperhomocysteinemia/metabolism , Male , Mice, Inbred C57BL , Mice, Mutant Strains , NAD(P)H Dehydrogenase (Quinone)/metabolism
15.
J Nutr ; 147(1): 11-19, 2017 01.
Article in English | MEDLINE | ID: mdl-27881594

ABSTRACT

BACKGROUND: Compared with choline, Met enhances milk yield and feed intake, and elicits a better immuno-metabolic status in periparturient cows. It is unknown whether hepatic activity and transcription of betaine-homocysteine methyltransferase (BHMT), 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR), and cystathionine ß-synthase (CBS) are responsive to Met and choline supply. OBJECTIVE: This study sought to characterize hepatic BHMT, MTR, and CBS transcription and activity in response to Met and choline supplementation. METHODS: Forty multiparous cows were used in a 2 × 2 factorial design from -21 d through 30 d around parturition to assess effects of dietary rumen-protected Met (0% or 0.08% dry matter basis) or rumen-protected choline (0 or 60 g · cow-1 · d-1). Liver tissue obtained on days -10, 7, 20, and 30 was used for analyses. RESULTS: Met-supplemented cows had greater methionine adenosyltransferase 1A (MAT1A) (0.38 compared with 0.27; SEM = 0.05; P = 0.02) and phosphatidylethanolamine methyltransferase (PEMT) (0.74 compared with 0.58; SEM = 0.08; P = 0.05) expression. Greater S-adenosylhomocysteine hydrolase (SAHH) (0.93 compared with 0.74; SEM = 0.05; P = 0.01) and CBS (1.16 compared with 1.02; SEM = 0.07; P = 0.04), as well as lower MTR activity (23.4 compared with 29.7 nmol product · h-1 · mg protein-1; SEM = 2.9; P = 0.04), also were detected in Met- but not choline-supplemented cows. Although BHMT and MTR expression and BHMT enzyme activity did not change (P > 0.05), MTR enzyme activity was lower in choline-supplemented cows (23.5 compared with 29.6 nmol product · h-1 · mg protein-1; SEM = 2.9; P = 0.05). CONCLUSIONS: These findings indicate that greater synthesis of phosphatidylcholine and antioxidants contribute to the better performance and immuno-metabolic status in Met-supplemented cows. Failure to generate a comparable amount of endogenous Met from choline could be one reason that choline-fed cows fail to achieve comparable performance and health benefits during the periparturient period.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Betaine-Homocysteine S-Methyltransferase/metabolism , Cattle/physiology , Choline/administration & dosage , Cystathionine beta-Synthase/metabolism , Methionine/administration & dosage , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Betaine-Homocysteine S-Methyltransferase/genetics , Cystathionine beta-Synthase/genetics , Diet/veterinary , Female , Gene Expression Regulation, Enzymologic , Liver/enzymology , Liver/metabolism , Peripartum Period
16.
Clin Chim Acta ; 458: 55-62, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27059523

ABSTRACT

Homocystinuria is a disorder of sulfur metabolism pathway caused by deficiency of cystathionine ß-synthase (CBS). It is characterized by increased accumulation of homocysteine (Hcy) in the cells and plasma. Increased homocysteine results in various vascular and neurological complications. Present strategies to lower cellular and plasma homocysteine levels include vitamin B6 intake, dietary methionine restriction, betaine supplementation, folate and vitamin B12 administration. However, these strategies are inefficient for treatment of homocystinuria. In recent years, advances have been made towards developing new strategies to treat homocystinuria. These mainly include functional restoration to mutant CBS, enhanced clearance of Hcy from the body, prevention of N-homocysteinylation-induced toxicity and inhibition of homocysteine-induced oxidative stress. In this review, we have exclusively discussed the recent advances that have been achieved towards the treatment of homocystinuria. The review is an attempt to help clinicians in developing effective therapeutic strategies and designing novel drugs against homocystinuria.


Subject(s)
Betaine/therapeutic use , Homocystinuria/drug therapy , Vitamin B 12/therapeutic use , Vitamin B 6/therapeutic use , Betaine/administration & dosage , Cystathionine beta-Synthase/deficiency , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Homocystinuria/enzymology , Humans , Hydrogen-Ion Concentration , Oxidative Stress/drug effects , Small Molecule Libraries/pharmacology , Vitamin B 12/administration & dosage , Vitamin B 6/administration & dosage
17.
Ann N Y Acad Sci ; 1363: 80-90, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26599618

ABSTRACT

Cystathionine ß synthase (CBS) is a key enzyme in the methionine and cysteine metabolic pathway, acting as a metabolic gatekeeper to regulate the flow of fixed sulfur from methionine to cysteine. Mutations in the CBS gene cause clinical CBS deficiency, a disease characterized by elevated plasma total homocysteine (tHcy) and methionine and decreased plasma cysteine. The treatment goal for CBS-deficient patients is to normalize the metabolic values of these three metabolites using a combination of vitamin therapy and dietary manipulation. To better understand the effectiveness of nutritional treatment strategies, we have performed a series of long-term dietary manipulation studies using our previously developed Tg-I278T Cbs(-/-) mouse model of CBS deficiency and sibling Tg-I278T Cbs(+/-) controls. Tg-I278T Cbs(-/-) mice have undetectable levels of CBS activity, extremely elevated plasma tHcy, modestly elevated plasma methionine, and low plasma cysteine. They exhibit several easily assayable phenotypes, including osteoporosis, loss of fat mass, reduced life span, and facial alopecia. The diets used in these studies differed in the amounts of sulfur amino acids or sulfur amino acid precursors. In this review, we will discuss our findings and their relevance to CBS deficiency and the concept of gene-diet interaction.


Subject(s)
Amino Acids, Sulfur/metabolism , Cystathionine beta-Synthase/deficiency , Diet , Homocystinuria/genetics , Homocystinuria/metabolism , Acetylcysteine/administration & dosage , Animals , Betaine/administration & dosage , Cystathionine beta-Synthase/genetics , Dietary Supplements , Disease Models, Animal , Genotype , Homocystinuria/diet therapy , Humans , Metabolic Networks and Pathways , Methionine/administration & dosage , Methionine/metabolism , Mice , Mice, Knockout , Mutation , Phenotype
18.
Sci Rep ; 5: 14971, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26455818

ABSTRACT

Oxidative stress and inflammation play crucial role in the pathogenesis of chronic obstructive pulmonary disease (COPD). Most patients with COPD show a poor response to corticosteroids. Hydrogen sulfide (H2S ) has been implicated in the pathogenesis of COPD, but its expression and effects in lung tissue from COPD patients are not clear. In peripheral lung tissue samples from 24 patients, we found that compared with nonsmokers, the protein level of cystathionine-γ-lyase (CSE) was decreased in smokers and COPD patients. CSE mRNA increased but cystathionine-ß-synthase (CBS) mRNA decreased in COPD patients. H2S donors increased glutathione and superoxide dismutase in CS exposed U937 cells and inhibited CS-induced TNF-α and IL-8 secretion. Dexamethasone alone had no effect on lipopolysaccharide (LPS) induced TNF-α release by alveolar macrophages from CS exposed rats, however the combination of dexamethasone and H2S donor significantly inhibited TNF-α release. Thus, H2S metabolism is altered in lung tissue of smokers and COPD patients. Supplementation of H2S protects against CS-induced oxidative stress and inflammation in macrophages and H2S on steroid sensitivity deserves further investigation.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Anti-Inflammatory Agents/pharmacology , Lung/metabolism , Macrophages/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , RNA, Messenger/metabolism , Animals , Cell Line, Tumor , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Dexamethasone/pharmacology , Gene Expression Regulation , Glutathione/metabolism , Humans , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Interleukin-8/genetics , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/pathology , Macrophages/drug effects , Macrophages/pathology , Oxidative Stress , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction , Smoking/adverse effects , Sulfides/metabolism , Sulfides/pharmacology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
19.
Cell Mol Neurobiol ; 35(6): 899-911, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25805165

ABSTRACT

Cystathionine-ß-synthase (CBS) deficiency is the main cause of homocystinuria. Homocysteine (Hcy), methionine, and other metabolites of Hcy accumulate in the body of affected patients. Despite the fact that thromboembolism represents the major cause of morbidity in CBS-deficient patients, the mechanisms of cardiovascular alterations found in homocystinuria remain unclear. In this work, we evaluated the lipid and inflammatory profile, oxidative protein damage, and the activities of the enzymes paraoxonase (PON1) and butyrylcholinesterase (BuChE) in plasma of CBS-deficient patients at diagnosis and during the treatment (protein-restricted diet supplemented with pyridoxine, folic acid, betaine, and vitamin B12). We also investigated the effect of folic acid and vitamin B12 on these parameters. We found a significant decrease in HDL cholesterol and apolipoprotein A1 (ApoA-1) levels, as well as in PON1 activity in both untreated and treated CBS-deficient patients when compared to controls. BuChE activity and IL-6 levels were significantly increased in not treated patients. Furthermore, significant positive correlations between PON1 activity and sulphydryl groups and between IL-6 levels and carbonyl content were verified. Moreover, vitamin B12 was positively correlated with PON1 and ApoA-1 levels, while folic acid was inversely correlated with total Hcy concentration, demonstrating the importance of this treatment. Our results also demonstrated that CBS-deficient patients presented important alterations in biochemical parameters, possibly caused by the metabolites of Hcy, as well as by oxidative stress, and that the adequate adherence to the treatment is essential to revert or prevent these alterations.


Subject(s)
Aryldialkylphosphatase/blood , Butyrylcholinesterase/blood , Homocystinuria/blood , Lipids/blood , Oxidants/blood , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Cystathionine beta-Synthase/deficiency , Cystathionine beta-Synthase/genetics , Female , Folic Acid/blood , Folic Acid/physiology , Homocystinuria/genetics , Humans , Male , Oxidative Stress/physiology , Vitamin B 12/blood , Vitamin B 12/physiology , Young Adult
20.
Clin Nutr ; 34(6): 1155-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25516282

ABSTRACT

BACKGROUND & AIMS: Many studies have reported that serum total homocysteine (tHcy) levels in cystathionine-beta-synthase (CBS) carriers are usually normal and only elevated after a methionine load. However, the amount of methionine required for a loading test is non-physiological and is never reached with regular feeding. Therefore, CBS carriers do not seem to be at an increased risk of cardiovascular diseases. However, the risk of cardiovascular diseases of CBS carriers with folate deficiency has not been studied. We recently found an extraordinarily high carrier rate (1/7.78) of a novel CBS mutation (p.D47E, c.T141A) in an Austronesian Taiwanese Tao tribe who live in a geographic area with folate deficiency. We evaluated if the CBS carriers tend to have higher fasting serum tHcy concentrations than non-carriers in presence of folate deficiency. METHODS: The serum tHcy and folate levels before and after folate replacement were measured in 48 adult Tao carriers, 40 age-matched Tao non-carriers and 40 age-matched Han Taiwanese controls. RESULTS: The serum tHcy level of the Tao CBS carriers (17.9 ± 3.8 µmol/l) was significantly higher than in Tao non-carriers (15.7 ± 3.5 µmol/l; p < 0.008) and Taiwanese controls (11.8 ± 2.9 µmol/l; p < 0.001). Furthermore, a high prevalence of folate deficiency in the Tao compared with the Taiwanese controls (4.9 ± 1.8 ng/ml vs. 10.6 ± 5.5 ng/ml; p < 0.001) was also noted. Of note, the difference in tHcy levels between the carriers and non-carriers was eliminated by folate supplementation. (carriers:13.65 ± 2.13 µmol/l; non-carriers:12.39 ± 3.25 µmol/l, p = 0.321). CONCLUSIONS: CBS carriers tend to have a higher tHcy level in the presence of folate deficiency than non-carriers. Although many reports have indicated that CBS carriers are not associated with cardiovascular disease, the risk for CBS carriers with folate deficiency has not been well studied. Owing to a significantly elevated level of fasting tHcy without methionine loading, it is important to evaluate the risk of cardiovascular disease in CBS carriers with folate deficiency.


Subject(s)
Folic Acid Deficiency/blood , Folic Acid Deficiency/epidemiology , Heterozygote , Homocysteine/blood , Homocystinuria/blood , Aged , Cardiovascular Diseases/blood , Case-Control Studies , Cystathionine beta-Synthase/blood , Cystathionine beta-Synthase/genetics , Dietary Supplements , Fasting , Female , Folic Acid/administration & dosage , Folic Acid/blood , Genotyping Techniques , Homocystinuria/genetics , Humans , Male , Methionine/administration & dosage , Methionine/blood , Methylenetetrahydrofolate Reductase (NADPH2)/blood , Middle Aged , Prevalence , Risk Factors , Taiwan , Vitamin B 12/blood
SELECTION OF CITATIONS
SEARCH DETAIL