Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Publication year range
1.
Angew Chem Int Ed Engl ; 63(20): e202402417, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38489608

ABSTRACT

Functionalizing single-walled carbon nanotubes (SWCNTs) in a robust way that does not affect the sp2 carbon framework is a considerable research challenge. Here we describe how triiodide salts of positively charged macrocycles can be used not only to functionalize SWCNTs from the outside, but simultaneously from the inside. We employed disulfide exchange in aqueous solvent to maximize the solvophobic effect and therefore achieve a high degree of macrocycle immobilization. Characterization by Raman spectroscopy, EDX-STEM and HR-TEM clearly showed that serendipitously this wet-chemical functionalization procedure also led to the encapsulation of polyiodide chains inside the nanotubes. The resulting three-shell composite materials are redox-active and experience an intriguing interplay of electrostatic, solvophobic and mechanical effects that could be of interest for applications in energy storage.

2.
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
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.
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
5.
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
6.
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
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.
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
9.
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
10.
Proc Natl Acad Sci U S A ; 110(40): E3790-9, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24043838

ABSTRACT

Cystathionine ß-synthase (CBS) controls the flux of sulfur from methionine to cysteine, a precursor of glutathione, taurine, and H2S. CBS condenses serine and homocysteine to cystathionine with the help of three cofactors, heme, pyridoxal-5'-phosphate, and S-adenosyl-l-methionine. Inherited deficiency of CBS activity causes homocystinuria, the most frequent disorder of sulfur metabolism. We present the structure of the human enzyme, discuss the unique arrangement of the CBS domains in the C-terminal region, and propose how they interact with the catalytic core of the complementary subunit to regulate access to the catalytic site. This arrangement clearly contrasts with other proteins containing the CBS domain including the recent Drosophila melanogaster CBS structure. The absence of large conformational changes and the crystal structure of the partially activated pathogenic D444N mutant suggest that the rotation of CBS motifs and relaxation of loops delineating the entrance to the catalytic site represent the most likely molecular mechanism of CBS activation by S-adenosyl-l-methionine. Moreover, our data suggest how tetramers, the native quaternary structure of the mammalian CBS enzymes, are formed. Because of its central role in transsulfuration, redox status, and H2S biogenesis, CBS represents a very attractive therapeutic target. The availability of the structure will help us understand the pathogenicity of the numerous missense mutations causing inherited homocystinuria and will allow the rational design of compounds modulating CBS activity.


Subject(s)
Cystathionine beta-Synthase/chemistry , Models, Molecular , Polymerization , Protein Conformation , Sulfur/chemistry , Blotting, Western , Crystallization , Denaturing Gradient Gel Electrophoresis , Humans
11.
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
12.
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
13.
Mol Ther ; 21(7): 1316-23, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23648696

ABSTRACT

Propionic acidemia (PA) is a recessive genetic disease that results in an inability to metabolize certain amino acids and odd-chain fatty acids. Current treatment involves restricting consumption of these substrates or liver transplantation. Deletion of the Pcca gene in mice mimics the most severe forms of the human disease. Pcca(-) mice die within 36 hours of birth, making it difficult to test intravenous systemic therapies in them. We generated an adult hypomorphic model of PA in Pcca(-) mice using a transgene bearing an A138T mutant of the human PCCA protein. Pcca(-/-)(A138T) mice have 2% of wild-type PCC activity, survive to adulthood, and have elevations in propionyl-carnitine, methylcitrate, glycine, alanine, lysine, ammonia, and markers associated with cardiomyopathy similar to those in patients with PA. This adult model allowed gene therapy testing by intravenous injection with adenovirus serotype 5 (Ad5) and adeno-associated virus 2/8 (AAV8) vectors. Ad5-mediated more rapid increases in PCCA protein and propionyl-CoA carboxylase (PCC) activity in the liver than AAV8 and both vectors reduced propionylcarnitine and methylcitrate levels. Phenotypic correction was transient with first generation Ad whereas AAV8-mediated long-lasting effects. These data suggest that this PA model may be a useful platform for optimizing systemic intravenous therapies for PA.


Subject(s)
Genetic Therapy/methods , Propionic Acidemia/therapy , Animals , Dependovirus/genetics , Disease Models, Animal , Humans , Methylmalonyl-CoA Decarboxylase/genetics , Methylmalonyl-CoA Decarboxylase/metabolism , Mice , Mice, Knockout , Mice, Transgenic
14.
Biochem J ; 449(1): 109-21, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22985361

ABSTRACT

CBS (cystathionine ß-synthase) is a multidomain tetrameric enzyme essential in the regulation of homocysteine metabolism, whose activity is enhanced by the allosteric regulator SAM (S-adenosylmethionine). Missense mutations in CBS are the major cause of inherited HCU (homocystinuria). In the present study we apply a novel approach based on a combination of calorimetric methods, functional assays and kinetic modelling to provide structural and energetic insight into the effects of SAM on the stability and activity of WT (wild-type) CBS and seven HCU-causing mutants. We found two sets of SAM-binding sites in the C-terminal regulatory domain with different structural and energetic features: a high affinity set of two sites, probably involved in kinetic stabilization of the regulatory domain, and a low affinity set of four sites, which are involved in the enzyme activation. We show that the regulatory domain displays a low kinetic stability in WT CBS, which is further decreased in many HCU-causing mutants. We propose that the SAM-induced stabilization may play a key role in modulating steady-state levels of WT and mutant CBS in vivo. Our strategy may be valuable for understanding ligand effects on proteins with a complex architecture and their role in human genetic diseases and for the development of novel pharmacological strategies.


Subject(s)
Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/pharmacokinetics , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/physiology , Binding Sites/genetics , Binding Sites/physiology , Cystathionine beta-Synthase/genetics , Enzyme Activation/genetics , Enzyme Activation/physiology , Homocystinuria/enzymology , Homocystinuria/genetics , Homocystinuria/metabolism , Humans , Protein Binding/genetics , Protein Stability , S-Adenosylmethionine/pharmacokinetics
15.
Biochemistry ; 52(4): 741-51, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23002992

ABSTRACT

Cystathionine ß-synthase (CBS) is the first and rate-limiting enzyme in the transsulfuration pathway, which is critical for the synthesis of cysteine from methionine in eukaryotes. CBS uses coenzyme pyridoxal 5'-phosphate (PLP) for catalysis, and S-adenosylmethionine regulates the activity of human CBS, but not yeast CBS. Human and fruit fly CBS contain heme; however, the role for heme is not clear. This paper reports biochemical and spectroscopic characterization of CBS from fruit fly Drosophila melanogaster (DmCBS) and the CO/NO gas binding reactions of DmCBS and human CBS. Like CBS enzymes from lower organisms (e.g., yeast), DmCBS is intrinsically highly active and is not regulated by AdoMet. The DmCBS heme coordination environment, the reactivity, and the accompanying effects on enzyme activity are similar to those of human CBS. The DmCBS heme bears histidine and cysteine axial ligands, and the enzyme becomes inactive when the cysteine ligand is replaced. The Fe(II) heme in DmCBS is less stable than that in human CBS, undergoing more facile reoxidation and ligand exchange. In both CBS proteins, the overall stability of the protein is correlated with the heme oxidation state. Human and DmCBS Fe(II) hemes react relatively slowly with CO and NO, and the rate of the CO binding reaction is faster at low pH than at high pH. Together, the results suggest that heme incorporation and AdoMet regulation in CBS are not correlated, possibly providing two independent means for regulating the enzyme.


Subject(s)
Cystathionine beta-Synthase/chemistry , Drosophila Proteins/chemistry , Drosophila melanogaster/enzymology , Heme/chemistry , Amino Acid Sequence , Animals , Carbon Monoxide/chemistry , Conserved Sequence , Cystathionine beta-Synthase/genetics , Drosophila Proteins/genetics , Enzyme Stability , Evolution, Molecular , Humans , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Nitric Oxide/chemistry , Oxidation-Reduction , Protein Binding
16.
PLoS Genet ; 6(1): e1000807, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20066033

ABSTRACT

Missense mutant proteins, such as those produced in individuals with genetic diseases, are often misfolded and subject to processing by intracellular quality control systems. Previously, we have shown using a yeast system that enzymatic function could be restored to I278T cystathionine beta-synthase (CBS), a cause of homocystinuria, by treatments that affect the intracellular chaperone environment. Here, we extend these studies and show that it is possible to restore significant levels of enzyme activity to 17 of 18 (94%) disease causing missense mutations in human cystathionine beta-synthase (CBS) expressed in Saccharomyces cerevisiae by exposure to ethanol, proteasome inhibitors, or deletion of the Hsp26 small heat shock protein. All three of these treatments induce Hsp70, which is necessary but not sufficient for rescue. In addition to CBS, these same treatments can rescue disease-causing mutations in human p53 and the methylene tetrahydrofolate reductase gene. These findings do not appear restricted to S. cerevisiae, as proteasome inhibitors can restore significant CBS enzymatic activity to CBS alleles expressed in fibroblasts derived from homocystinuric patients and in a mouse model for homocystinuria that expresses human I278T CBS. These findings suggest that proteasome inhibitors and other Hsp70 inducing agents may be useful in the treatment of a variety of genetic diseases caused by missense mutations.


Subject(s)
Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Enzyme Inhibitors/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Homocystinuria/metabolism , Mutation , Proteasome Inhibitors , Animals , Boronic Acids/pharmacology , Bortezomib , Cell Line , Cells, Cultured , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , HSP70 Heat-Shock Proteins/genetics , Homocystinuria/drug therapy , Homocystinuria/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyrazines/pharmacology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
17.
Angew Chem Int Ed Engl ; 52(17): 4641-4, 2013 Apr 22.
Article in English | MEDLINE | ID: mdl-23512751

ABSTRACT

Buzzing with activity: A hydrogen sulfide selective fluorogenic probe, 7-azido-4-methylcoumarin (AzMC), serves as a highly sensitive assay for cystathionine ß-synthase activity, and is suitable for the high-throughput discovery of novel enzyme inhibitors.


Subject(s)
Cystathionine beta-Synthase/antagonists & inhibitors , Cystathionine beta-Synthase/chemistry , Enzyme Inhibitors/chemistry , Fluorescent Dyes/chemistry , Hydrogen Sulfide/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays
18.
Biochemistry ; 51(32): 6360-70, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22738154

ABSTRACT

Cystathionine ß-synthase (CBS) is an essential pyridoxal 5'-phosphate (PLP)-dependent enzyme of the transsulfuration pathway that condenses serine with homocysteine to form cystathionine; intriguingly, human CBS also contains a heme b cofactor of unknown function. Herein we describe the enzymatic and spectroscopic properties of a disease-associated R266K hCBS variant, which has an altered hydrogen-bonding environment. The R266K hCBS contains a low-spin, six-coordinate Fe(III) heme bearing a His/Cys ligation motif, like that of WT hCBS; however, there is a geometric distortion that exists at the R266K heme. Using rR spectroscopy, we show that the Fe(III)-Cys(thiolate) bond is longer and weaker in R266K, as evidenced by an 8 cm(-1) downshift in the ν(Fe-S) resonance. Presence of this longer and weaker Fe(III)-Cys(thiolate) bond is correlated with alteration of the fluorescence spectrum of the active PLP ketoenamine tautomer. Activity data demonstrate that, relative to WT, the R266K variant is more impaired in the alternative cysteine-synthesis reaction than in the canonical cystathionine-synthesis reaction. This diminished cysteine synthesis activity and a greater sensitivity to exogenous PLP correlate with the change in PLP environment. Fe-S(Cys) bond weakening causes a nearly 300-fold increase in the rate of ligand switching upon reduction of the R266K heme. Combined, these data demonstrate cross talk between the heme and PLP active sites, consistent with previous proposals, revealing that alteration of the Arg(266)-Cys(52) interaction affects PLP-dependent activity and dramatically destabilizes the ferrous thiolate-ligated heme complex, underscoring the importance of this hydrogen-bonding residue pair.


Subject(s)
Cystathionine beta-Synthase/chemistry , Heme/genetics , Pyridoxal Phosphate/chemistry , Catalytic Domain , Circular Dichroism , Cystathionine beta-Synthase/genetics , Electron Spin Resonance Spectroscopy , Enzyme Stability , Ferrous Compounds/chemistry , Homocystinuria/genetics , Humans , Models, Molecular , Mutation , Oxidation-Reduction , Protein Binding , Spectrometry, Fluorescence , Spectrum Analysis, Raman , Temperature
19.
Biochemistry ; 51(23): 4755-63, 2012 06 12.
Article in English | MEDLINE | ID: mdl-22612060

ABSTRACT

Protein misfolding due to missense mutations is a common pathogenic mechanism in cystathionine ß-synthase (CBS) deficiency. In our previous studies, we successfully expressed, purified, and characterized nine CBS mutant enzymes containing the following patient mutations: P49L, P78R, A114V, R125Q, E176K, R266K, P422L, I435T, and S466L. These purified mutants exhibited full heme saturation, normal tetrameric assembly, and high catalytic activity. In this work, we used several spectroscopic and proteolytic techniques to provide a more thorough insight into the conformation of these mutant enzymes. Far-UV circular dichroism, fluorescence, and second-derivative UV spectroscopy revealed that the spatial arrangement of these CBS mutants is similar to that of the wild type, although the microenvironment of the chromophores may be slightly altered. Using proteolysis with thermolysin under native conditions, we found that the majority of the studied mutants is more susceptible to cleavage, suggesting their increased local flexibility or propensity for local unfolding. Interestingly, the presence of the CBS allosteric activator, S-adenosylmethionine (AdoMet), increased the rate of cleavage of the wild type and the AdoMet-responsive mutants, while the proteolytic rate of the AdoMet-unresponsive mutants was not significantly changed. Pulse proteolysis analysis suggested that the protein structure of the R125Q and E176K mutants is significantly less stable than that of the wild type and the other mutants. Taken together, the proteolytic data shows that the conformation of the pathogenic mutants is altered despite retained catalytic activity and normal tetrameric assembly. This study demonstrates that the proteolytic techniques are useful tools for the assessment of the biochemical penalty of missense mutations in CBS.


Subject(s)
Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Protein Folding , Circular Dichroism , Cystathionine beta-Synthase/deficiency , Escherichia coli/metabolism , Humans , Models, Molecular , Mutation, Missense , Protein Conformation , Proteolysis , S-Adenosylmethionine , Spectrophotometry, Ultraviolet
20.
Protein Expr Purif ; 82(2): 317-24, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22333527

ABSTRACT

Cystathionine ß-synthase (CBS), a heme-containing pyridoxal-5-phosphate (PLP)-dependent enzyme, catalyzes the condensation of serine and homocysteine to yield cystathionine. Missense mutations in CBS, the most common cause of homocystinuria, often result in misfolded proteins. Arginine 266, where the pathogenic missense mutation R266K was identified, appears to be involved in the communication between heme and the PLP-containing catalytic center. Here, we assessed the effect of a short affinity tag (6xHis) compared to a bulky fusion partner (glutathione S-transferase - GST) on CBS wild type (WT) and R266K mutant enzyme properties. While WT CBS was successfully expressed either in conjunction with a GST or with a 6xHis tag, the mutant R266K CBS had no activity, did not form native tetramers and did not respond to chemical chaperone treatment when expressed with a GST fusion partner. Interestingly, expression of R266K CBS constructs with a 6xHis tag at either end yielded active enzymes. The purified, predominantly tetrameric, R266K CBS with a C-terminal 6xHis tag had ∼82% of the activity of a corresponding WT CBS construct. Results from thermal pre-treatment of the enzyme and the denaturation profile of R266K suggests a lower thermal stability of the mutant enzyme compared to WT, presumably due to a disturbed heme environment.


Subject(s)
Cystathionine beta-Synthase/chemistry , Mutant Proteins/chemistry , Protein Folding , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Amino Acid Substitution , Cloning, Molecular , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/isolation & purification , Enzyme Stability , Hot Temperature , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Transition Temperature
SELECTION OF CITATIONS
SEARCH DETAIL