Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Inherit Metab Dis ; 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38873792

RESUMEN

This review summarises progress in the research of homocystinuria (HCU) in the past three decades. HCU due to cystathionine ß-synthase (CBS) was discovered in 1962, and Prof. Jan Peter Kraus summarised developments in the field in the first-ever Komrower lecture in 1993. In the past three decades, significant advancements have been achieved in the biology of CBS, including gene organisation, tissue expression, 3D structures, and regulatory mechanisms. Renewed interest in CBS arose in the late 1990s when this enzyme was implicated in biogenesis of H2S. Advancements in genetic and biochemical techniques enabled the identification of several hundreds of pathogenic CBS variants and the misfolding of missense mutations as a common mechanism. Several cellular, invertebrate and murine HCU models allowed us to gain insights into functional and metabolic pathophysiology of the disease. Establishing the E-HOD consortium and patient networks, HCU Network Australia and HCU Network America, offered new possibilities for acquiring clinical data in registries and data on patients´ quality of life. A recent analysis of data from the E-HOD registry showed that the clinical variability of HCU is broad, extending from severe childhood disease to milder (late) adulthood forms, which typically respond to pyridoxine. Pyridoxine responsiveness appears to be the key factor determining the clinical course of HCU. Increased awareness about HCU played a role in developing novel therapies, such as gene therapy, correction of misfolding by chaperones, removal of methionine from the gut and enzyme therapies that decrease homocysteine or methionine in the circulation.

2.
Cell Mol Life Sci ; 79(8): 438, 2022 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-35864237

RESUMEN

Cystathionine beta-synthase (CBS) is a pivotal enzyme of the transsulfuration pathway responsible for diverting homocysteine to the biosynthesis of cysteine and production of hydrogen sulfide (H2S). Aberrant upregulation of CBS and overproduction of H2S contribute to pathophysiology of several diseases including cancer and Down syndrome. Therefore, pharmacological CBS inhibition has emerged as a prospective therapeutic approach. Here, we characterized binding and inhibitory mechanism of aminooxyacetic acid (AOAA), the most commonly used CBS inhibitor. We found that AOAA binds CBS tighter than its respective substrates and forms a dead-end PLP-bound intermediate featuring an oxime bond. Surprisingly, serine, but not cysteine, replaced AOAA from CBS and formed an aminoacrylate reaction intermediate, which allowed for the continuation of the catalytic cycle. Indeed, serine rescued and essentially normalized the enzymatic activity of AOAA-inhibited CBS. Cellular studies confirmed that AOAA decreased H2S production and bioenergetics, while additional serine rescued CBS activity, H2S production and mitochondrial function. The crystal structure of AOAA-bound human CBS showed a lack of hydrogen bonding with residues G305 and Y308, found in the serine-bound model. Thus, AOAA-inhibited CBS could be reactivated by serine. This difference may be important in a cellular environment in multiple pathophysiological conditions and may modulate the CBS-inhibitory activity of AOAA. In addition, our results demonstrate additional complexities of using AOAA as a CBS-specific inhibitor of H2S biogenesis and point to the urgent need to develop a potent, selective and specific pharmacological CBS inhibitor.


Asunto(s)
Cistationina betasintasa , Sulfuro de Hidrógeno , Ácido Aminooxiacético/farmacología , Cistationina betasintasa/metabolismo , Cisteína , Humanos , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/farmacología , Serina
3.
Nitric Oxide ; 128: 12-24, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35973674

RESUMEN

Epigallocatechin gallate (EGCG) is the main bioactive component of green tea. Through screening of a small library of natural compounds, we discovered that EGCG inhibits cystathionine ß-synthase (CBS), a major H2S-generating enzyme. Here we characterize EGCG's mechanism of action in the context of CBS-derived H2S production. In the current project, biochemical, pharmacological and cell biology approaches were used to characterize the effect of EGCG on CBS in cellular models of cancer and Down syndrome (DS). The results show that EGCG binds to CBS and inhibits H2S-producing CBS activity almost 30-times more efficiently than the canonical cystathionine formation (IC50 0.12 versus 3.3 µM). Through screening structural analogs and building blocks, we identified that gallate moiety of EGCG represents the pharmacophore responsible for CBS inhibition. EGCG is a mixed-mode, CBS-specific inhibitor with no effect on the other two major enzymatic sources of H2S, CSE and 3-MST. Unlike the prototypical CBS inhibitor aminooxyacetate, EGCG does not bind the catalytic cofactor of CBS pyridoxal-5'-phosphate. Molecular modeling suggests that EGCG blocks a substrate access channel to pyridoxal-5'-phosphate. EGCG inhibits cellular H2S production in HCT-116 colon cancer cells and in DS fibroblasts. It also exerts effects that are consistent with the functional role of CBS in these cells: in HCT-116 cells it decreases, while in DS cells it improves viability and proliferation. In conclusion, EGCG is a potent inhibitor of CBS-derived H2S production. This effect may contribute to its pharmacological effects in various pathophysiological conditions.


Asunto(s)
Cistationina betasintasa , Sulfuro de Hidrógeno , Catequina/análogos & derivados , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Humanos , Sulfuro de Hidrógeno/metabolismo , Fosfatos , Piridoxal , Relación Estructura-Actividad
4.
Curr Opin Clin Nutr Metab Care ; 24(1): 62-70, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33060459

RESUMEN

PURPOSE OF REVIEW: Metabolism of sulfur amino acids (SAA) provides compounds important for many cellular functions. Inherited disorders of SAA metabolism are typically severe multisystemic diseases affecting brain, liver, connective tissue, or vasculature. The review summarizes the present therapeutic approaches and advances in identifying novel treatment targets, and provides an overview of new therapies. RECENT FINDINGS: Current treatments of genetic disorders of SAA metabolism are primarily based on modulation of affected pathways by dietary measures and provision of lacking products or scavenging of toxic molecules. Recent studies identified additional therapeutic targets distant from the primary defects and explored ideas envisioning novel treatments, such as chaperone and gene therapy. Recombinant protein production and engineering resulted in development and clinical testing of enzyme therapies for cystathionine ß-synthase deficiency, the most common inborn error of SAA metabolism. SUMMARY: Complex regulation of pathways involved in SAA metabolism and cellular consequences of genetic defects in SAA metabolism are only partially understood. There is a pressing need to increase substantially our knowledge of the disease mechanisms to develop more effective therapies for patients suffering from these rare disorders.


Asunto(s)
Aminoácidos Sulfúricos , Homocistinuria , Encéfalo , Dieta , Humanos , Hígado
5.
Hum Mutat ; 41(9): 1662-1670, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32623804

RESUMEN

Classical homocystinuria (HCU) is an inborn error of metabolism caused by loss of cystathionine ß-synthase (CBS) activity with the concomitant buildup of homocysteine. In knockout (KO) mice, a mouse model of HCU, complete lack of CBS is neonatally lethal. Administration of OT-58, an enzyme therapy for HCU, during the first 5 weeks of life rescued KO mice survival by preventing liver disease. Here, we studied the impact of a long-term uninterrupted OT-58 treatment or its absence beyond the neonatal period on liver pathology and metabolism. Plasma and liver metabolites of KO mice on OT-58 treatment were substantially improved or normalized compared with those receiving vehicle. Increased plasma activities of alanine aminotransferase and aspartate aminotransferase of vehicle-injected KO mice suggested the progression of liver damage with age and lack of treatment. At 3 months of age, liver histology showed no signs of hepatopathy in both vehicle- and OT-58-treated KO mice. However, moderate to severe liver disease, characterized by steatosis, hepatocellular necroses, disorganized endoplasmic reticulum, and swollen mitochondria, developed in 6-month-old vehicle-injected KO mice. KO mice on OT-58 treatment remained asymptomatic and were indistinguishable from age-matched healthy controls. Long-term uninterrupted OT-58 treatment was essential to prevent severe liver disease in the KO mouse model of HCU.


Asunto(s)
Terapia de Reemplazo Enzimático , Homocistinuria/tratamiento farmacológico , Hepatopatías/prevención & control , Animales , Modelos Animales de Enfermedad , Femenino , Hígado/patología , Masculino , Ratones , Ratones Noqueados
6.
FASEB J ; 33(11): 12477-12486, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31450979

RESUMEN

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.


Asunto(s)
Conducta Animal , Composición Corporal , Cistationina betasintasa/uso terapéutico , Terapia de Reemplazo Enzimático , Homocistinuria/tratamiento farmacológico , Animales , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Modelos Animales de Enfermedad , Homocistinuria/genética , Homocistinuria/metabolismo , Homocistinuria/patología , Humanos , Metionina/farmacología , Ratones , Ratones Transgénicos
7.
Mol Ther ; 26(3): 834-844, 2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29398487

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/administración & dosificación , Terapia de Reemplazo Enzimático , Homocistinuria/diagnóstico , Homocistinuria/terapia , Fenotipo , Aminoácidos Sulfúricos/sangre , Aminoácidos Sulfúricos/metabolismo , Animales , Cistationina betasintasa/química , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Glucosa/metabolismo , Homocistinuria/metabolismo , Metabolismo de los Lípidos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Estrés Oxidativo , Polietilenglicoles/química
8.
Hum Mutat ; 39(2): 210-218, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29044829

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/uso terapéutico , Terapia de Reemplazo Enzimático/métodos , Homocistinuria/tratamiento farmacológico , Absorciometría de Fotón , Animales , Composición Corporal , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Femenino , Homocistinuria/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
J Struct Biol ; 202(1): 82-93, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29275181

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/química , Proteínas de Insectos/química , Conformación Proteica , Multimerización de Proteína , Secuencia de Aminoácidos , Animales , Abejas , Cristalografía por Rayos X , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Cisteína/metabolismo , Homocisteína/metabolismo , Humanos , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Modelos Moleculares , S-Adenosilmetionina/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
10.
FASEB J ; 31(12): 5495-5506, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28821635

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/metabolismo , Cistationina betasintasa/uso terapéutico , Hígado Graso/prevención & control , Homocistinuria/tratamiento farmacológico , Homocistinuria/enzimología , Hepatopatías/prevención & control , Osteoporosis/prevención & control , Animales , Composición Corporal/efectos de los fármacos , Cistationina betasintasa/genética , Modelos Animales de Enfermedad , Hígado Graso/enzimología , Femenino , Homocistinuria/metabolismo , Homocistinuria/patología , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/metabolismo , Hígado/patología , Hepatopatías/enzimología , Masculino , Ratones , Ratones Noqueados , Proteínas Recombinantes/uso terapéutico
11.
Handb Exp Pharmacol ; 245: 345-383, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29119254

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/deficiencia , Homocistinuria/tratamiento farmacológico , Chaperonas Moleculares/uso terapéutico , Animales , Cistationina betasintasa/química , Cistationina betasintasa/fisiología , Estabilidad de Enzimas , Ensayos Analíticos de Alto Rendimiento , Humanos , Pliegue de Proteína , Procesamiento Proteico-Postraduccional
12.
Biomacromolecules ; 18(6): 1747-1761, 2017 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-28431470

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/química , Cistationina betasintasa/farmacocinética , Preparaciones de Acción Retardada/síntesis química , Terapia de Reemplazo Enzimático/métodos , Homocistinuria/terapia , Polietilenglicoles/química , Succinimidas/química , Secuencia de Aminoácidos , Animales , Reactivos de Enlaces Cruzados/química , Cistationina betasintasa/farmacología , Cisteína/sangre , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Modelos Animales de Enfermedad , Homocisteína/sangre , Homocistinuria/sangre , Homocistinuria/fisiopatología , Humanos , Maleimidas/química , Ratones
13.
Proc Natl Acad Sci U S A ; 111(37): E3845-52, 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25197074

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/química , Cistationina betasintasa/metabolismo , S-Adenosilmetionina/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Activación Enzimática , Estabilidad de Enzimas , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína
14.
Proc Natl Acad Sci U S A ; 110(40): E3790-9, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24043838

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/química , Modelos Moleculares , Polimerizacion , Conformación Proteica , Azufre/química , Western Blotting , Cristalización , Electroforesis en Gel de Gradiente Desnaturalizante , Humanos
15.
Biochim Biophys Acta ; 1844(9): 1453-62, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24780582

RESUMEN

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.


Asunto(s)
Adenosina/análogos & derivados , Cistationina betasintasa/química , Etionina/análogos & derivados , Subunidades de Proteína/química , Adenosina/química , Adenosina/metabolismo , Dominio Catalítico , Cistationina betasintasa/metabolismo , Activación Enzimática , Etionina/química , Etionina/metabolismo , Humanos , Cinética , Ligandos , Unión Proteica , Multimerización de Proteína , Estabilidad Proteica , Estructura Secundaria de Proteína , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Cloruro de Sodio/química , Electricidad Estática , Propiedades de Superficie , Termodinámica , Urea/química
16.
Bioorg Med Chem Lett ; 25(5): 1064-6, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25666819

RESUMEN

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.


Asunto(s)
Aminas/química , Cistationina betasintasa/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Sulfuro de Hidrógeno/metabolismo , Imidazoles/química , Urocordados/química , Aminas/aislamiento & purificación , Aminas/farmacología , Animales , Productos Biológicos/química , Productos Biológicos/aislamiento & purificación , Productos Biológicos/farmacología , Cistationina betasintasa/metabolismo , Inhibidores Enzimáticos/aislamiento & purificación , Inhibidores Enzimáticos/farmacología , Humanos , Sulfuro de Hidrógeno/análisis , Imidazoles/aislamiento & purificación , Imidazoles/farmacología
17.
Biochem J ; 449(1): 109-21, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22985361

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/química , Cistationina betasintasa/farmacocinética , S-Adenosilmetionina/química , S-Adenosilmetionina/fisiología , Sitios de Unión/genética , Sitios de Unión/fisiología , Cistationina betasintasa/genética , Activación Enzimática/genética , Activación Enzimática/fisiología , Homocistinuria/enzimología , Homocistinuria/genética , Homocistinuria/metabolismo , Humanos , Unión Proteica/genética , Estabilidad Proteica , S-Adenosilmetionina/farmacocinética
18.
Protein Sci ; 33(8): e5123, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39041895

RESUMEN

Homocystinuria (HCU) due to cystathionine beta-synthase (CBS) deficiency is the most common inborn error of sulfur amino acid metabolism. Recent work suggests that missense pathogenic mutations-regardless of their topology-cause instability of the C-terminal regulatory domain, which likely translates into CBS misfolding, impaired assembly, and loss of function. However, it is unknown how instability of the regulatory domain translates into cellular CBS turnover and which degradation pathways are involved in CBS proteostasis. Here, we developed a human HEK293-based cellular model lacking intrinsic CBS and stably overexpressing wild-type (WT) CBS or its 10 most common missense HCU mutants. We found that HCU mutants, except the I278T variant, expressed similarly or better than CBS WT, with some of them showing impaired oligomerization, activity and response to allosteric activator S-adenosylmethionine. Cellular stability of all HCU mutants, except P49L and A114V, was significantly lower than the stability of CBS WT, suggesting their increased degradation. Ubiquitination analysis of CBS WT and two representative CBS mutants (T191M and I278T) showed that proteasomal degradation is the major pathway for CBS disposal, with a minor involvement of lysosomal-autophagic and endoplasmic reticulum-associated degradation (ERAD) pathways for HCU mutants. Proteasomal inhibition significantly increased the half-life and activity of T191M and I278T CBS mutants. Lysosomal and ERAD inhibition had only a minor impact on CBS turnover, but ERAD inhibition rescued the activity of T191M and I278T CBS mutants similarly as proteasomal inhibition. In conclusion, the present study provides new insights into proteostasis of CBS in HCU.


Asunto(s)
Cistationina betasintasa , Homocistinuria , Mutación Missense , Proteolisis , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Cistationina betasintasa/química , Humanos , Homocistinuria/genética , Homocistinuria/metabolismo , Células HEK293 , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Ubiquitinación , Degradación Asociada con el Retículo Endoplásmico
19.
Redox Biol ; 73: 103222, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38843767

RESUMEN

BACKGROUND: Cystathionine ß-synthase (CBS)-deficient homocystinuria (HCU) is an inherited disorder of sulfur amino acid metabolism with varying severity and organ complications, and a limited knowledge about underlying pathophysiological processes. Here we aimed at getting an in-depth insight into disease mechanisms using a transgenic mouse model of HCU (I278T). METHODS: We assessed metabolic, proteomic and sphingolipidomic changes, and mitochondrial function in tissues and body fluids of I278T mice and WT controls. Furthermore, we evaluated the efficacy of methionine-restricted diet (MRD) in I278T mice. RESULTS: In WT mice, we observed a distinct tissue/body fluid compartmentalization of metabolites with up to six-orders of magnitude differences in concentrations among various organs. The I278T mice exhibited the anticipated metabolic imbalance with signs of an increased production of hydrogen sulfide and disturbed persulfidation of free aminothiols. HCU resulted in a significant dysregulation of liver proteome affecting biological oxidations, conjugation of compounds, and metabolism of amino acids, vitamins, cofactors and lipids. Liver sphingolipidomics indicated upregulation of the pro-proliferative sphingosine-1-phosphate signaling pathway. Liver mitochondrial function of HCU mice did not seem to be impaired compared to controls. MRD in I278T mice improved metabolic balance in all tissues and substantially reduced dysregulation of liver proteome. CONCLUSION: The study highlights distinct tissue compartmentalization of sulfur-related metabolites in normal mice, extensive metabolome, proteome and sphingolipidome disruptions in I278T mice, and the efficacy of MRD to alleviate some of the HCU-related biochemical abnormalities.


Asunto(s)
Cistationina betasintasa , Modelos Animales de Enfermedad , Homocistinuria , Hígado , Metabolómica , Ratones Transgénicos , Proteómica , Esfingolípidos , Animales , Ratones , Homocistinuria/metabolismo , Homocistinuria/genética , Proteómica/métodos , Cistationina betasintasa/metabolismo , Cistationina betasintasa/deficiencia , Cistationina betasintasa/genética , Hígado/metabolismo , Metabolómica/métodos , Esfingolípidos/metabolismo , Mitocondrias/metabolismo , Lipidómica/métodos , Proteoma/metabolismo
20.
Sci Rep ; 14(1): 9364, 2024 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-38654065

RESUMEN

The escalating drug resistance among microorganisms underscores the urgent need for innovative therapeutic strategies and a comprehensive understanding of bacteria's defense mechanisms against oxidative stress and antibiotics. Among the recently discovered barriers, the endogenous production of hydrogen sulfide (H2S) via the reverse transsulfuration pathway, emerges as a noteworthy factor. In this study, we have explored the catalytic capabilities and crystal structure of cystathionine γ-lyase from Pseudomonas aeruginosa (PaCGL), a multidrug-opportunistic pathogen chiefly responsible for nosocomial infections. In addition to a canonical L-cystathionine hydrolysis, PaCGL efficiently catalyzes the production of H2S using L-cysteine and/or L-homocysteine as alternative substrates. Comparative analysis with the human enzyme and counterparts from other pathogens revealed distinct structural features within the primary enzyme cavities. Specifically, a distinctly folded entrance loop could potentially modulate the access of substrates and/or inhibitors to the catalytic site. Our findings offer significant insights into the structural evolution of CGL enzymes across different pathogens and provide novel opportunities for developing specific inhibitors targeting PaCGL.


Asunto(s)
Dominio Catalítico , Cistationina gamma-Liasa , Sulfuro de Hidrógeno , Pseudomonas aeruginosa , Pseudomonas aeruginosa/enzimología , Cistationina gamma-Liasa/metabolismo , Cistationina gamma-Liasa/química , Cristalografía por Rayos X , Especificidad por Sustrato , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/química , Modelos Moleculares , Cisteína/metabolismo , Cisteína/química , Conformación Proteica , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Humanos , Homocisteína/metabolismo , Homocisteína/química , Catálisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA