Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
FASEB J ; 33(11): 12477-12486, 2019 11.
Article in English | MEDLINE | ID: mdl-31450979

ABSTRACT

Classic homocystinuria (HCU) is an inherited disorder characterized by elevated homocysteine (Hcy) in plasma and tissues resulting from cystathionine ß-synthase (CBS) deficiency. There is no cure, and patients are predominantly managed by methionine-restricted diet (MRD) to limit the production of Hcy. In this study, we used the I278T mouse model of HCU to evaluate the long-term impact of a novel enzyme replacement therapy [truncated human CBS C15S mutant modified with linear 20-kDa N-hydroxysuccinimide ester polyethylene glycol (OT-58)] on clinical end points relevant to human patients with HCU. In addition, we compared its efficacy on a background of either MRD or normal methionine intake [regular diet (REG)] to that of MRD alone. We found that, compared with untreated I278T mice, OT-58 treatment of I278T mice fed with the REG diet resulted in a 90% decrease in plasma Hcy concentrations and correction of learning/cognition, endothelial dysfunction, hemostasis, bone mineralization, and body composition. On background of the MRD, OT-58 performed equally well with plasma Hcy entirely normalized. The MRD alone decreased plasma Hcy by 67% and corrected the HCU phenotype in I278T mice. However, the MRD increased anxiety and reduced bone mineral content in both I278T mice and wild-type controls. This study shows that OT-58 is a highly efficacious novel treatment for HCU on the background of either normal or restricted methionine intake.-Majtan, T., Park, I., Cox, A., Branchford, B. R., di Paola, J., Bublil, E. M., Kraus, J. P. Behavior, body composition, and vascular phenotype of homocystinuric mice on methionine-restricted diet or enzyme replacement therapy.


Subject(s)
Behavior, Animal , Body Composition , Cystathionine beta-Synthase/therapeutic use , Enzyme Replacement Therapy , Homocystinuria/drug therapy , Animals , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Disease Models, Animal , Homocystinuria/genetics , Homocystinuria/metabolism , Homocystinuria/pathology , Humans , Methionine/pharmacology , Mice , Mice, Transgenic
2.
Life Sci ; 200: 15-25, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29526799

ABSTRACT

AIMS: PEGylated human truncated cystathionine beta-synthase, lacking the C-terminal regulatory domain (PEG-CBS), is a promising preclinical candidate for enzyme replacement therapy in homocystinuria (HCU). It was designed to function as a metabolic sink to decrease the severely elevated plasma and tissue homocysteine concentrations. In this communication, we evaluated pharmacokinetics (PK), pharmacodynamics (PD) and sub-chronic toxicity of PEG-CBS in homocystinuric mice, wild type rats and monkeys to estimate the minimum human efficacious dose for clinical trials. MAIN METHODS: Animal models received single or multiple doses of PEG-CBS. Activity of PEG-CBS and sulfur amino acid metabolites were determined in plasma and used to determine PK and PD. KEY FINDINGS: The plasma half-lives of PEG-CBS after a single subcutaneous (SC) injection were approximately 20, 44 and 73 h in mouse, rat and monkey, respectively. The SC administration of PEG-CBS resulted in a significant improvement or full correction of metabolic imbalance in both blood and tissues of homocystinuric mice. The PD of PEG-CBS in mouse was dose-dependent, but less than dose-proportional, with the maximal efficacy achieved at 8 mg/kg. PEG-CBS was well-tolerated in mice and monkeys, but resulted in dose-dependent minimal-to-moderate inflammation at the injection sites and vacuolated macrophages in rats. Allometric scaling of animal data was linear and the estimated human efficacious dose was determined as 0.66 mg/kg administered once a week. SIGNIFICANCE: These results provide critical preclinical data for the design of first-in-human PEG-CBS clinical trial.


Subject(s)
Cystathionine beta-Synthase/pharmacokinetics , Cystathionine beta-Synthase/therapeutic use , Enzyme Replacement Therapy , Homocystinuria/drug therapy , Animals , Cystathionine beta-Synthase/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Homocystinuria/genetics , Homocystinuria/metabolism , Humans , Macaca fascicularis , Male , Mice , Mice, Knockout , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/therapeutic use , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use
3.
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
4.
Mol Cell Biol ; 38(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29378828

ABSTRACT

Propionic acidemia is caused by a deficiency of the enzyme propionyl coenzyme A carboxylase (PCC) located in the mitochondrial matrix. Cell-penetrating peptides, including transactivator of transcription (TAT), offer a potential to deliver a cargo into the mitochondrion. Here, we investigated the delivery of an α6ß6 PCC enzyme into mitochondria using the HIV TAT peptide at several levels: into isolated mitochondria, in patient fibroblast cells, and in a mouse model. Results from Western blots and enzyme activity assays confirmed the import of TAT-PCC into mitochondria, as well as into patient fibroblasts, where the colocalization of imported TAT-PCC and mitochondria was also confirmed by confocal fluorescence microscopy. Furthermore, a single-dose intraperitoneal injection into PCC-deficient mice decreased the propionylcarnitine/acetylcarnitine (C3/C2) ratio toward the normal level. These results show that a cell-penetrating peptide can deliver active multimeric enzyme into mitochondria in vitro, in situ, and in vivo and push the size limit of intracellular delivery achieved so far. Our results are promising for other mitochondrion-specific deficiencies.


Subject(s)
Methylmalonyl-CoA Decarboxylase/administration & dosage , Methylmalonyl-CoA Decarboxylase/therapeutic use , Nanoconjugates/administration & dosage , Nanoconjugates/therapeutic use , Propionic Acidemia/drug therapy , tat Gene Products, Human Immunodeficiency Virus/chemistry , Animals , Carnitine/analogs & derivatives , Carnitine/metabolism , Cell-Penetrating Peptides/chemistry , Cells, Cultured , Humans , Methylmalonyl-CoA Decarboxylase/chemistry , Methylmalonyl-CoA Decarboxylase/pharmacokinetics , Mice , Mitochondria/metabolism , Nanoconjugates/chemistry , Propionic Acidemia/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use
5.
Antioxid Redox Signal ; 28(4): 311-323, 2018 02 01.
Article in English | MEDLINE | ID: mdl-28874062

ABSTRACT

AIMS: The transsulfuration pathway enzymes cystathionine beta-synthase (CBS) and cystathionine gamma-lyase are thought to be the major source of hydrogen sulfide (H2S). In this study, we assessed the role of CBS in H2S biogenesis. RESULTS: We show that despite discouraging enzyme kinetics of alternative H2S-producing reactions utilizing cysteine compared with the canonical condensation of serine and homocysteine, our simulations of substrate competitions at biologically relevant conditions suggest that cysteine is able to partially compete with serine on CBS, thus leading to generation of appreciable amounts of H2S. The leading H2S-producing reaction is condensation of cysteine with homocysteine, while cysteine desulfuration plays a dominant role when cysteine is more abundant than serine and homocysteine is limited. We found that the serine-to-cysteine ratio is the main determinant of CBS H2S productivity. Abundance of cysteine over serine, for example, in plasma, allowed for up to 43% of CBS activity being responsible for H2S production, while excess of serine typical for intracellular levels effectively limited such activity to less than 1.5%. CBS also produced lanthionine from serine and cysteine and a third of lanthionine coming from condensation of two cysteines contributed to the H2S pool. INNOVATION: Our study characterizes the H2S-producing potential of CBS under biologically relevant conditions and highlights the serine-to-cysteine ratio as the main determinant of H2S production by CBS in vivo. CONCLUSION: Our data clarify the function of CBS in H2S biogenesis and the role of thioethers as surrogate H2S markers. Antioxid. Redox Signal. 28, 311-323.


Subject(s)
Biomarkers/metabolism , Cystathionine beta-Synthase/metabolism , Hydrogen Sulfide/metabolism , Sulfides/metabolism , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/metabolism , Animals , Biomarkers/chemistry , Catalysis , Cystathionine beta-Synthase/chemistry , Cysteine/chemistry , Haplorhini , Homocysteine/chemistry , Hydrogen Sulfide/chemistry , Kinetics , Mice , Mice, Knockout , Serine/chemistry , Sulfides/chemistry , Sulfur/metabolism
6.
J Struct Biol ; 202(1): 82-93, 2018 04.
Article in English | MEDLINE | ID: mdl-29275181

ABSTRACT

Cystathionine ß-synthase (CBS), the key enzyme in the transsulfuration pathway, links methionine metabolism to the biosynthesis of cellular redox controlling molecules. CBS catalyzes the pyridoxal-5'-phosphate-dependent condensation of serine and homocysteine to form cystathionine, which is subsequently converted into cysteine. Besides maintaining cellular sulfur amino acid homeostasis, CBS also catalyzes multiple hydrogen sulfide-generating reactions using cysteine and homocysteine as substrates. In mammals, CBS is activated by S-adenosylmethionine (AdoMet), where it can adopt two different conformations (basal and activated), but exists as a unique highly active species in fruit fly Drosophila melanogaster. Here we present the crystal structure of CBS from honeybey Apis mellifera, which shows a constitutively active dimeric species and let explain why the enzyme is not allosterically regulated by AdoMet. In addition, comparison of available CBS structures unveils a substrate-induced closure of the catalytic cavity, which in humans is affected by the AdoMet-dependent regulation and likely impaired by the homocystinuria causing mutation T191M.


Subject(s)
Cystathionine beta-Synthase/chemistry , Insect Proteins/chemistry , Protein Conformation , Protein Multimerization , Amino Acid Sequence , Animals , Bees , Crystallography, X-Ray , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cysteine/metabolism , Homocysteine/metabolism , Humans , Insect Proteins/genetics , Insect Proteins/metabolism , Models, Molecular , S-Adenosylmethionine/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
7.
Handb Exp Pharmacol ; 245: 345-383, 2018.
Article in English | MEDLINE | ID: mdl-29119254

ABSTRACT

Classical homocystinuria (HCU) is the most common loss-of-function inborn error of sulfur amino acid metabolism. HCU is caused by a deficiency in enzymatic degradation of homocysteine, a toxic intermediate of methionine transformation to cysteine, chiefly due to missense mutations in the cystathionine beta-synthase (CBS) gene. As with many other inherited disorders, the pathogenic mutations do not target key catalytic residues, but rather introduce structural perturbations leading to an enhanced tendency of the mutant CBS to misfold and either to form nonfunctional aggregates or to undergo proteasome-dependent degradation. Correction of CBS misfolding would represent an alternative therapeutic approach for HCU. In this review, we summarize the complex nature of CBS, its multi-domain architecture, the interplay between the three cofactors required for CBS function [heme, pyridoxal-5'-phosphate (PLP), and S-adenosylmethionine (SAM)], as well as the intricate allosteric regulatory mechanism only recently understood, thanks to advances in CBS crystallography. While roughly half of the patients respond to treatment with a PLP precursor pyridoxine, many studies suggested usefulness of small chemicals, such as chemical and pharmacological chaperones or proteasome inhibitors, rescuing mutant CBS activity in cellular and animal models of HCU. Non-specific chemical chaperones and proteasome inhibitors assist in mutant CBS folding process and/or prevent its rapid degradation, thus resulting in increased steady-state levels of the enzyme and CBS activity. Recent interest in the field and available structural information will hopefully yield CBS-specific compounds, by using high-throughput screening and computational modeling of novel ligands, improving folding, stability, and activity of CBS mutants.


Subject(s)
Cystathionine beta-Synthase/deficiency , Homocystinuria/drug therapy , Molecular Chaperones/therapeutic use , Animals , Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/physiology , Enzyme Stability , High-Throughput Screening Assays , Humans , Protein Folding , Protein Processing, Post-Translational
8.
Hum Mutat ; 39(2): 210-218, 2018 02.
Article in English | MEDLINE | ID: mdl-29044829

ABSTRACT

Skeletal and connective tissue defects are the most striking symptoms in patients suffering from classical homocystinuria (HCU). Here, we determined body composition and bone mass in three mouse models of HCU and assessed whether a long-term administration of enzyme replacement therapy (ERT) corrected the phenotype. The mouse models of HCU were analyzed using dual-energy X-ray absorptiometry and the data were complemented by plasma biochemical profiles. Both the mouse model lacking CBS (KO) and the one expressing human CBS mutant transgene on a mouse CBS null background (I278T) showed marked bone loss and decreased weight mostly due to a lower fat content compared with negative controls. In contrast, the HO mouse expressing the human CBS WT transgene on a mouse CBS null background showed no such phenotype despite similar plasma biochemical profile to the KO and I278T mice. More importantly, administration of ERT rescued bone mass and changes in body composition in the KO mice treated since birth and reversed bone loss and improved fat content in the I278T mice injected after the development of clinical symptoms. Our study suggests that ERT for HCU may represent an effective way of preventing the skeletal problems in patients without a restricted dietary regime.


Subject(s)
Cystathionine beta-Synthase/therapeutic use , Enzyme Replacement Therapy/methods , Homocystinuria/drug therapy , Absorptiometry, Photon , Animals , Body Composition , Bone Diseases, Metabolic/drug therapy , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Female , Homocystinuria/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
FASEB J ; 31(12): 5495-5506, 2017 12.
Article in English | MEDLINE | ID: mdl-28821635

ABSTRACT

Classical homocystinuria (HCU) is an inborn error of sulfur amino acid metabolism caused by deficient activity of cystathionine ß-synthase (CBS), resulting in an accumulation of homocysteine and a concomitant decrease of cystathionine and cysteine in blood and tissues. In mice, the complete lack of CBS is neonatally lethal. In this study, newborn CBS-knockout (KO) mice were treated with recombinant polyethyleneglycolylated human truncated CBS (PEG-CBS). Full survival of the treated KO mice, along with a positive impact on metabolite levels in plasma, liver, brain, and kidneys, was observed. The PEG-CBS treatment prevented an otherwise fatal liver disease characterized by steatosis, death of hepatocytes, and ultrastructural abnormalities of endoplasmic reticulum and mitochondria. Furthermore, treatment of the KO mice for 5 mo maintained the plasma metabolite balance and completely prevented osteoporosis and changes in body composition that characterize both the KO model and human patients. These findings argue that early treatment of patients with HCU with PEG-CBS may prevent clinical symptoms of the disease possibly without the need of dietary protein restriction.-Majtan, T., Hulková, H., Park, I., Krijt, J., Kozich, V., Bublil, E. M., Kraus, J. P. Enzyme replacement prevents neonatal death, liver damage, and osteoporosis in murine homocystinuria.


Subject(s)
Cystathionine beta-Synthase/metabolism , Cystathionine beta-Synthase/therapeutic use , Fatty Liver/prevention & control , Homocystinuria/drug therapy , Homocystinuria/enzymology , Liver Diseases/prevention & control , Osteoporosis/prevention & control , Animals , Body Composition/drug effects , Cystathionine beta-Synthase/genetics , Disease Models, Animal , Fatty Liver/enzymology , Female , Homocystinuria/metabolism , Homocystinuria/pathology , Liver/drug effects , Liver/enzymology , Liver/metabolism , Liver/pathology , Liver Diseases/enzymology , Male , Mice , Mice, Knockout , Recombinant Proteins/therapeutic use
10.
Biomacromolecules ; 18(6): 1747-1761, 2017 Jun 12.
Article in English | MEDLINE | ID: mdl-28431470

ABSTRACT

Homocystinuria due to loss of cystathionine beta-synthase (CBS) causes accumulation of homocysteine and depletion of cysteine. Current treatments are suboptimal, and thus the development of an enzyme replacement therapy based on PEGylated human truncated CBS (PEG-CBS) has been initiated. Attenuation of potency was observed, which necessitated a screen of several PEG-CBS conjugates for their efficacy to correct and maintain the plasma metabolite profile of murine homocystinuria after repeated administrations interrupted with washouts. We found that CBS coupling with maleimide PEG inconsistently modified the enzyme. In contrast, the PEG-CBS conjugate with 20 kDa N-hydroxysuccinimide-PEG showed very little loss of potency likely due to a reproducible PEGylation resulting in species modified with five PEGs per subunit on average. We developed assays suitable for monitoring the extent of CBS PEGylation and demonstrated a sustainable partial normalization of homocystinuria upon continuous PEG-CBS administration via osmotic pumps. Taken together, we identified the PEG-CBS conjugate suitable for manufacturing and clinical development.


Subject(s)
Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/pharmacokinetics , Delayed-Action Preparations/chemical synthesis , Enzyme Replacement Therapy/methods , Homocystinuria/therapy , Polyethylene Glycols/chemistry , Succinimides/chemistry , Amino Acid Sequence , Animals , Cross-Linking Reagents/chemistry , Cystathionine beta-Synthase/pharmacology , Cysteine/blood , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Disease Models, Animal , Homocysteine/blood , Homocystinuria/blood , Homocystinuria/physiopathology , Humans , Maleimides/chemistry , Mice
11.
FEBS Lett ; 590(24): 4461-4471, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27861796

ABSTRACT

Cystathionine beta-synthase (CBS) plays a key role in the metabolism of sulfur-containing amino acids. CBS is a multidomain tetrameric enzyme allosterically activated by S-adenosylmethionine (AdoMet). Recent crystallographic analyses of engineered CBS lacking the loop made up of residues 516-525 revealed discrepancies in AdoMet binding compared to previous biophysical studies on a full-length CBS. Here, we show that removal of the loop 516-525 functionally eliminates the high affinity sites responsible for kinetic stabilization of the full-length enzyme and yields a dimeric AdoMet-inducible enzyme, in which kinetic stabilization is now exerted by AdoMet binding to the remaining low affinity sites.


Subject(s)
Cystathionine beta-Synthase/chemistry , Recombinant Fusion Proteins/chemistry , S-Adenosylmethionine/chemistry , Allosteric Regulation , Binding Sites , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Enzyme Stability , Gene Expression , Humans , Kinetics , Mutation , Protein Binding , Protein Domains , Protein Multimerization , Protein Structure, Secondary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , S-Adenosylmethionine/metabolism , Thermodynamics
12.
J Clin Invest ; 126(6): 2372-84, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183385

ABSTRACT

Homocystinuria, which typically results from cystathionine ß-synthase (CBS) deficiency, is the most common defect of sulfur amino acid metabolism. CBS condenses homocysteine and serine to cystathionine that is then converted to cysteine. Individuals with homocystinuria have markedly elevated plasma levels of homocysteine and methionine and reduced concentrations of cystathionine and cysteine. Clinical disease manifestations include thromboembolism and neuropsychiatric, ocular, and skeletal complications. Here, we have shown that administration of PEGylated CBS into the circulation of homocystinuria model mice alters the extra- and intracellular equilibrium of sulfur amino acids, resulting in a decrease of approximately 75% in plasma total homocysteine (tHcy) and normalization of cysteine concentrations. Moreover, the decrease in homocysteine and the normalization of cysteine in PEGylated CBS-treated model mice were accompanied by improvement of histopathological liver symptoms and increased survival. Together, these data suggest that CBS enzyme replacement therapy (ERT) is a promising approach for the treatment of homocystinuria and that ERT for metabolic diseases may not necessitate introduction of the deficient enzyme into its natural intracellular compartment.


Subject(s)
Cystathionine beta-Synthase/deficiency , Cystathionine beta-Synthase/therapeutic use , Homocystinuria/drug therapy , Homocystinuria/metabolism , Animals , Cystathionine beta-Synthase/genetics , Disease Models, Animal , Homocystinuria/pathology , Humans , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyethylene Glycols , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use
13.
Curr Drug Targets ; 17(13): 1455-70, 2016.
Article in English | MEDLINE | ID: mdl-26931358

ABSTRACT

Classical homocystinuria (HCU) is the most common loss-of-function inborn error of sulfur amino acids metabolism. HCU is caused by a deficiency in enzymatic degradation of homocysteine, a toxic intermediate of methionine transformation to cysteine, chiefly due to missense mutations in the cystathionine betasynthase (CBS) gene. As with many other inherited disorders, the pathogenic mutations do not target key catalytic residues, but rather introduce structural perturbations leading to an enhanced tendency of the mutant CBS to misfold and either to form non-functional aggregates or to undergo proteasome-dependent degradation. Thus correction of CBS misfolding represents an alternative therapeutic approach for HCU. In this review, we summarize the complex nature of CBS, its multidomain architecture, the interplay between the three cofactors required for CBS function (heme, pyridoxal-5'-phosphate (PLP) and S-adenosyl-L-methionine) as well as the intricate allosteric regulatory mechanism only recently explained thanks to advances in CBS crystallography. While roughly half of the patients responds to treatment with a PLP precursor pyridoxine, many studies suggested usefulness of small chemicals, such as chemical and pharmacological chaperones or proteasome inhibitors, rescuing mutant CBS activity in cellular and animal models of HCU. Non-specific chemical chaperones and proteasome inhibitors assist in mutant CBS folding process and/or prevent its rapid degradation, thus resulting in increased steady state levels of the enzyme and CBS activity. Recent increased interest in the field and available structural information will hopefully yield CBS-specific compounds by using high-throughput screening and computational modeling of novel ligands improving folding, stability and activity of CBS.


Subject(s)
Cystathionine beta-Synthase/metabolism , Homocystinuria/drug therapy , Molecular Targeted Therapy , Animals , Drug Design , High-Throughput Screening Assays , Homocystinuria/physiopathology , Humans , Ligands , Molecular Chaperones/pharmacology , Proteasome Inhibitors/pharmacology , Protein Folding/drug effects
14.
Biochimie ; 126: 14-20, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26791043

ABSTRACT

Two enzymes in the transsulfuration pathway of homocysteine -cystathionine beta-synthase (CBS) and gamma-cystathionase (CTH)-use cysteine and/or homocysteine to produce the important signaling molecule hydrogen sulfide (H2S) and simultaneously the thioethers lanthionine, cystathionine or homolanthionine. In this study we explored whether impaired flux of substrates for H2S synthesis and/or deficient enzyme activities alter production of hydrogen sulfide in patients with homocystinurias. As an indirect measure of H2S synthesis we determined by LC-MS/MS concentrations of thioethers in plasma samples from 33 patients with different types of homocystinurias, in 8 patient derived fibroblast cell lines, and as reaction products of seven purified mutant CBS enzymes. Since chaperoned recombinant mutant CBS enzymes retained capacity of H2S synthesis in vitro it can be stipulated that deficient CBS activity in vivo may impair H2S production. Indeed, in patients with classical homocystinuria we observed significantly decreased cystathionine and lanthionine concentrations in plasma (46% and 74% of median control levels, respectively) and significantly lower cystathionine in fibroblasts (8% of median control concentrations) indicating that H2S production from cysteine and homocysteine may be also impaired. In contrast, the grossly elevated plasma levels of homolanthionine in CBS deficient patients (32-times elevation compared to median of controls) clearly demonstrates a simultaneous overproduction of H2S from homocysteine by CTH. In the remethylation defects the accumulation of homocysteine and the increased flux of metabolites through the transsulfuration pathway resulted in elevation of cystathionine and homolanthionine (857% and 400% of median control values, respectively) indicating a possibility of an increased biosynthesis of H2S by both CBS and CTH. This study shows clearly disturbed thioether concentrations in homocystinurias, and modeling using these data indicates that H2S synthesis may be increased in these conditions. Further studies are needed to confirm our findings and to explore the possible implications for pathophysiology of these disorders.


Subject(s)
Alanine/analogs & derivatives , Cystathionine/metabolism , Fibroblasts/metabolism , Homocystinuria/metabolism , Hydrogen Sulfide/metabolism , Sulfides/metabolism , Alanine/metabolism , Cells, Cultured , Cystathionine beta-Synthase/metabolism , Female , Fibroblasts/pathology , Homocystinuria/pathology , Humans , Male
15.
Biochimie ; 126: 6-13, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26805382

ABSTRACT

Many pathogenic missense mutations in human cystathionine beta-synthase (CBS) cause misfolding of the mutant enzyme resulting in aggregation or rapid degradation of the protein. Subsequent loss of CBS function leads to CBS-deficient homocystinuria (CBSDH). CBS contains two sets of binding sites for S-adenosylmethionine (SAM) that independently regulate the enzyme activity and kinetically stabilize its regulatory domain. In the present study, we examined the hypothesis that CBS activation may be decoupled from kinetic stabilization and thus CBS regulatory domain can serve as a novel drug target for CBSDH. We determined the effect of SAM and its close structural analogs on CBS activity, their binding to and stabilization of the regulatory domain in the absence and presence of competing SAM. Binding of S-adenosylhomocysteine and sinefungin lead to stabilization of the regulatory domains without activation of CBS. Direct titrations and competition experiments support specific binding of these two SAM analogs to the stabilizing sites. Binding of these two ligands also affects the enzyme proteolysis rate supporting the role of the stabilizing sites in CBS dynamics. Our results indicate that binding of SAM to regulatory and stabilizing sites in CBS may have evolved to display an exquisite thermodynamic and structural specificity towards SAM as well as the ability to transduce the allosteric signal responsible for CBS activation. Thus, ligands may be developed to function as kinetic stabilizers or pharmacological chaperones without interfering with the physiological activation of CBS by SAM.


Subject(s)
Cystathionine beta-Synthase/metabolism , Homocystinuria/drug therapy , Homocystinuria/enzymology , S-Adenosylmethionine/analogs & derivatives , S-Adenosylmethionine/pharmacology , Enzyme Stability/drug effects , Humans , Kinetics , S-Adenosylmethionine/chemistry
16.
Hum Gene Ther ; 26(3): 153-60, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25654275

ABSTRACT

Propionic academia (PA) occurs because of mutations in the PCCA or PCCB genes encoding the two subunits of propionyl-CoA carboxylase, a pivotal enzyme in the breakdown of certain amino acids and odd-chain fatty acids. There is no cure for PA, but dietary protein restriction and liver transplantation can attenuate its symptoms. We show here that a single intravenous injection of adeno-associated virus 2/8 (AAV8) or AAVrh10 expressing PCCA into PA hypomorphic mice decreased systemic propionylcarnitine and methyl citrate for up to 1.5 years. However, long-term phenotypic correction was always better in male mice. AAV-mediated PCCA expression was similar in most tissues in males and females at early time points and differed only in the liver. Over 1.5 years, luciferase and PCCA expression remained elevated in cardiac tissue for both sexes. In contrast, transgene expression in the liver and skeletal muscles of female, but not male, mice waned­suggesting that these tissues were major sinks for systemic phenotypic correction. These data indicate that single systemic intravenous therapy by AAV vectors can mediate long-term phenotype correction for PA. However, tissue-specific loss of expression in females reduces efficacy when compared with males. Whether similar sex-biased AAV effects occur in human gene therapy remains to be determined.


Subject(s)
Biomarkers/blood , Genetic Therapy/methods , Genetic Vectors/genetics , Methylmalonyl-CoA Decarboxylase/metabolism , Propionic Acidemia/genetics , Propionic Acidemia/therapy , Sex Characteristics , Animals , Carnitine/analogs & derivatives , Carnitine/blood , Citrates/blood , Dependovirus , Female , Injections, Intravenous , Liver/metabolism , Luciferases , Male , Methylmalonyl-CoA Decarboxylase/genetics , Mice , Muscle, Skeletal/metabolism , Propionic Acidemia/blood , Real-Time Polymerase Chain Reaction
17.
Bioorg Med Chem Lett ; 25(5): 1064-6, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25666819

ABSTRACT

A library consisting of characterized marine natural products as well as synthetic derivatives was screened for compounds capable of inhibiting the production of hydrogen sulfide (H2S) by cystathionine beta-synthase (CBS). Eight hits were validated and shown to inhibit CBS activity with IC50 values ranging from 83 to 187µM. The majority of hits came from a series of synthetic polyandrocarpamine derivatives. In addition, a modified fluorogenic probe for H2S detection with improved solubility in aqueous solutions is reported.


Subject(s)
Amines/chemistry , Cystathionine beta-Synthase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Hydrogen Sulfide/metabolism , Imidazoles/chemistry , Urochordata/chemistry , Amines/isolation & purification , Amines/pharmacology , Animals , Biological Products/chemistry , Biological Products/isolation & purification , Biological Products/pharmacology , Cystathionine beta-Synthase/metabolism , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Sulfide/analysis , Imidazoles/isolation & purification , Imidazoles/pharmacology
18.
Proc Natl Acad Sci U S A ; 111(37): E3845-52, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25197074

ABSTRACT

Cystathionine ß-synthase (CBS) is a heme-dependent and pyridoxal-5'-phosphate-dependent protein that controls the flux of sulfur from methionine to cysteine, a precursor of glutathione, taurine, and H2S. Deficiency of CBS activity causes homocystinuria, the most frequent disorder of sulfur amino acid metabolism. In contrast to CBSs from lower organisms, human CBS (hCBS) is allosterically activated by S-adenosylmethionine (AdoMet), which binds to the regulatory domain and triggers a conformational change that allows the protein to progress from the basal toward the activated state. The structural basis of the underlying molecular mechanism has remained elusive so far. Here, we present the structure of hCBS with bound AdoMet, revealing the activated conformation of the human enzyme. Binding of AdoMet triggers a conformational change in the Bateman module of the regulatory domain that favors its association with a Bateman module of the complementary subunit to form an antiparallel CBS module. Such an arrangement is very similar to that found in the constitutively activated insect CBS. In the presence of AdoMet, the autoinhibition exerted by the regulatory region is eliminated, allowing for improved access of substrates to the catalytic pocket. Based on the availability of both the basal and the activated structures, we discuss the mechanism of hCBS activation by AdoMet and the properties of the AdoMet binding site, as well as the responsiveness of the enzyme to its allosteric regulator. The structure described herein paves the way for the rational design of compounds modulating hCBS activity and thus transsulfuration, redox status, and H2S biogenesis.


Subject(s)
Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/metabolism , S-Adenosylmethionine/metabolism , Allosteric Regulation , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Enzyme Activation , Enzyme Stability , Humans , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Structure, Secondary
19.
PLoS One ; 9(8): e105290, 2014.
Article in English | MEDLINE | ID: mdl-25122507

ABSTRACT

Cystathionine beta-synthase (CBS) is a key regulator of sulfur amino acid metabolism diverting homocysteine, a toxic intermediate of the methionine cycle, via the transsulfuration pathway to the biosynthesis of cysteine. Although the pathway itself is well conserved among eukaryotes, properties of eukaryotic CBS enzymes vary greatly. Here we present a side-by-side biochemical and biophysical comparison of human (hCBS), fruit fly (dCBS) and yeast (yCBS) enzymes. Preparation and characterization of the full-length and truncated enzymes, lacking the regulatory domains, suggested that eukaryotic CBS exists in one of at least two significantly different conformations impacting the enzyme's catalytic activity, oligomeric status and regulation. Truncation of hCBS and yCBS, but not dCBS, resulted in enzyme activation and formation of dimers compared to native tetramers. The dCBS and yCBS are not regulated by the allosteric activator of hCBS, S-adenosylmethionine (AdoMet); however, they have significantly higher specific activities in the canonical as well as alternative reactions compared to hCBS. Unlike yCBS, the heme-containing dCBS and hCBS showed increased thermal stability and retention of the enzyme's catalytic activity. The mass-spectrometry analysis and isothermal titration calorimetry showed clear presence and binding of AdoMet to yCBS and hCBS, but not dCBS. However, the role of AdoMet binding to yCBS remains unclear, unlike its role in hCBS. This study provides valuable information for understanding the complexity of the domain organization, catalytic specificity and regulation among eukaryotic CBS enzymes.


Subject(s)
Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/metabolism , Protein Interaction Domains and Motifs , Animals , Catalysis , Catalytic Domain , Cystathionine beta-Synthase/genetics , Enzyme Activation , Gene Expression , Humans , Models, Molecular , Protein Conformation , Protein Denaturation , Protein Stability , Recombinant Proteins , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thermodynamics
20.
Biochim Biophys Acta ; 1844(9): 1453-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24780582

ABSTRACT

Human cystathionine ß-synthase (hCBS) is a key enzyme of sulfur amino acid metabolism, controlling the commitment of homocysteine to the transsulfuration pathway and antioxidant defense. Mutations in hCBS cause inherited homocystinuria (HCU), a rare inborn error of metabolism characterized by accumulation of toxic homocysteine in blood and urine. hCBS is a complex multidomain and oligomeric protein whose activity and stability are independently regulated by the binding of S-adenosyl-methionine (SAM) to two different types of sites at its C-terminal regulatory domain. Here we study the role of surface electrostatics on the complex regulation and stability of hCBS using biophysical and biochemical procedures. We show that the kinetic stability of the catalytic and regulatory domains is significantly affected by the modulation of surface electrostatics through noticeable structural and energetic changes along their denaturation pathways. We also show that surface electrostatics strongly affect SAM binding properties to those sites responsible for either enzyme activation or kinetic stabilization. Our results provide new insight into the regulation of hCBS activity and stability in vivo with implications for understanding HCU as a conformational disease. We also lend experimental support to the role of electrostatic interactions in the recently proposed binding modes of SAM leading to hCBS activation and kinetic stabilization.


Subject(s)
Adenosine/analogs & derivatives , Cystathionine beta-Synthase/chemistry , Ethionine/analogs & derivatives , Protein Subunits/chemistry , Adenosine/chemistry , Adenosine/metabolism , Catalytic Domain , Cystathionine beta-Synthase/metabolism , Enzyme Activation , Ethionine/chemistry , Ethionine/metabolism , Humans , Kinetics , Ligands , Protein Binding , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sodium Chloride/chemistry , Static Electricity , Surface Properties , Thermodynamics , Urea/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...