Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
1.
Biochem J ; 481(8): 569-585, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38563463

RESUMEN

Homocystinuria is a rare disease caused by mutations in the CBS gene that results in a deficiency of cystathionine ß-synthase (CBS). CBS is an essential pyridoxal 5'-phosphate (PLP)-dependent enzyme in the transsulfuration pathway, responsible for combining serine with homocysteine to produce cystathionine, whose activity is enhanced by the allosteric regulator S-adenosylmethionine (SAM). CBS also plays a role in generating hydrogen sulfide (H2S), a gaseous signaling molecule with diverse regulatory functions within the vascular, nervous, and immune systems. In this study, we present the clinical and biochemical characterization of two novel CBS missense mutations that do not respond to pyridoxine treatment, namely c.689T > A (L230Q) and 215A > T (K72I), identified in a Chinese patient. We observed that the disease-associated K72I genetic variant had no apparent effects on the spectroscopic and catalytic properties of the full-length enzyme. In contrast, the L230Q variant expressed in Escherichia coli did not fully retain heme and when compared with the wild-type enzyme, it exhibited more significant impairments in both the canonical cystathionine-synthesis and the alternative H2S-producing reactions. This reduced activity is consistent with both in vitro and in silico evidence, which indicates that the L230Q mutation significantly decreases the overall protein's stability, which in turn, may represent the underlying cause of its pathogenicity.


Asunto(s)
Cistationina betasintasa , Homocistinuria , Mutación Missense , Cistationina betasintasa/genética , Cistationina betasintasa/química , Cistationina betasintasa/metabolismo , Homocistinuria/genética , Homocistinuria/metabolismo , Homocistinuria/enzimología , Humanos , Masculino , Femenino
2.
J Biol Chem ; 299(12): 105449, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37949228

RESUMEN

Cystathionine ß-synthase (CBS) catalyzes the committing step in the transsulfuration pathway, which is important for clearing homocysteine and furnishing cysteine. The transsulfuration pathway also generates H2S, a signaling molecule. CBS is a modular protein with a heme and pyridoxal phosphate-binding catalytic core, which is separated by a linker region from the C-terminal regulatory domain that binds S-adenosylmethionine (AdoMet), an allosteric activator. Recent cryo-EM structures reveal that CBS exists in a fibrillar form and undergoes a dramatic architectural rearrangement between the basal and AdoMet-bound states. CBS is the single most common locus of mutations associated with homocystinuria, and, in this study, we have characterized three clinical variants (K384E/N and M391I), which reside in the linker region. The native fibrillar form is destabilized in the variants, and differences in their limited proteolytic fingerprints also reveal conformational alterations. The crystal structure of the truncated K384N variant, lacking the regulatory domain, reveals that the overall fold of the catalytic core is unperturbed. M391I CBS exhibits a modest (1.4-fold) decrease while the K384E/N variants exhibit a significant (∼8-fold) decrease in basal activity, which is either unresponsive to or inhibited by AdoMet. Pre-steady state kinetic analyses reveal that the K384E/N substitutions exhibit pleiotropic effects and that the differences between them are expressed in the second half reaction, that is, homocysteine binding and reaction with the aminoacrylate intermediate. Together, these studies point to an important role for the linker in stabilizing the higher-order oligomeric structure of CBS and enabling AdoMet-dependent regulation.


Asunto(s)
Cistationina betasintasa , Mutación , Humanos , Regulación Alostérica/genética , Cristalografía por Rayos X , Cistationina betasintasa/química , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Homocisteína/metabolismo , Homocistinuria/enzimología , Homocistinuria/genética , Cinética , S-Adenosilmetionina/metabolismo , Conformación Proteica , Dominio Catalítico
3.
Curr Eye Res ; 46(4): 600-605, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32865440

RESUMEN

PURPOSE: Deficiency in Cystathionine ß-synthase (CBS) leads to an abnormal accumulation of homocysteine and results in classical homocystinuria, a multi-systemic disorder that affects connective tissue, muscles, the central nervous system, and the eyes. However, the genetic players and mechanisms underlying vision alterations in patients with homocystinuria are little understood. MATERIALS AND METHODS: The fruit fly, Drosophila melanogaster, is a useful system to investigate the genetic basis of several human diseases, but no study to date has used Drosophila as model of homocystinuria. Here, we use Drosophila genetic tools to down-regulate CBS expression and evaluate its behavioral response to light. RESULTS: We show that CBS-deficient flies do not display the normal stereotypical behavior of attraction towards a luminous source, known as phototaxis. This behavior cannot be attributed to a motor or olfactory deficiency, but it is most likely related to a lower visual acuity. CBS-deficient flies are overall smaller, but smaller eyes do not explain their lack of phototactic response. CONCLUSIONS: The vision phenotype of CBS knock-down flies is consistent with severe myopia in homocystinuria patients. We propose to use Drosophila as a model to investigate ocular manifestations underlying homocystinuria.


Asunto(s)
Cistationina betasintasa/deficiencia , Drosophila melanogaster/enzimología , Fototaxis/fisiología , Trastornos de la Visión/enzimología , Animales , Western Blotting , Cistationina betasintasa/genética , Modelos Animales de Enfermedad , Drosophila melanogaster/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Homocisteína/metabolismo , Homocistinuria/enzimología , Trastornos de la Visión/fisiopatología
4.
J Inherit Metab Dis ; 44(3): 677-692, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33295057

RESUMEN

Cystathionine ß-synthase (CBS) deficiency has a wide clinical spectrum, ranging from neurodevelopmental problems, lens dislocation and marfanoid features in early childhood to adult onset disease with predominantly thromboembolic complications. We have analysed clinical and laboratory data at the time of diagnosis in 328 patients with CBS deficiency from the E-HOD (European network and registry for Homocystinurias and methylation Defects) registry. We developed comprehensive criteria to classify patients into four groups of pyridoxine responsivity: non-responders (NR), partial, full and extreme responders (PR, FR and ER, respectively). All groups showed overlapping concentrations of plasma total homocysteine while pyridoxine responsiveness inversely correlated with plasma/serum methionine concentrations. The FR and ER groups had a later age of onset and diagnosis and a longer diagnostic delay than NR and PR patients. Lens dislocation was common in all groups except ER but the age of dislocation increased with increasing responsiveness. Developmental delay was commonest in the NR group while no ER patient had cognitive impairment. Thromboembolism was the commonest presenting feature in ER patients, whereas it was least likely at presentation in the NR group. This probably is due to the differences in ages at presentation: all groups had a similar number of thromboembolic events per 1000 patient-years. Clinical severity of CBS deficiency depends on the degree of pyridoxine responsiveness. Therefore, a standardised pyridoxine-responsiveness test in newly diagnosed patients and a critical review of previous assessments is indispensable to ensure adequate therapy and to prevent or reduce long-term complications.


Asunto(s)
Cistationina betasintasa/deficiencia , Homocistinuria/diagnóstico , Homocistinuria/tratamiento farmacológico , Piridoxina/uso terapéutico , Adolescente , Adulto , Anciano , Niño , Preescolar , Diagnóstico Tardío , Europa (Continente) , Femenino , Homocistinuria/enzimología , Humanos , Lactante , Modelos Lineales , Masculino , Metionina/sangre , Persona de Mediana Edad , Fenotipo , Sistema de Registros , Índice de Severidad de la Enfermedad , Adulto Joven
5.
J Am Heart Assoc ; 9(4): e013368, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32067580

RESUMEN

Background Hyperhomocysteinemia is a risk factor for ischemic stroke; however, a targeted treatment strategy is lacking partly because of limited understanding of the causal role of homocysteine in cerebrovascular pathogenesis. Methods and Results In a genetic model of cystathionine beta synthase (CBS) deficiency, we tested the hypothesis that elevation in plasma total homocysteine exacerbates cerebrovascular injury and that memantine, a N-methyl-D-aspartate receptor antagonist, is protective. Mild or severe elevation in plasma total homocysteine was observed in Cbs+/- (6.1±0.3 µmol/L) or Cbs-/- (309±18 µmol/L) mice versus Cbs+/+ (3.1±0.6 µmol/L) mice. Surprisingly, Cbs-/- and Cbs+/- mice exhibited similar increases in cerebral infarct size following middle cerebral artery ischemia/reperfusion injury, despite the much higher total homocysteine levels in Cbs-/- mice. Likewise, disruption of the blood brain barrier was observed in both Cbs+/- and Cbs-/- mice. Administration of the N-methyl-D-aspartate receptor antagonist memantine protected Cbs+/- but not Cbs-/- mice from cerebral infarction and blood brain barrier disruption. Our data suggest that the differential effect of memantine in Cbs+/- versus Cbs-/- mice may be related to changes in expression of N-methyl-D-aspartate receptor subunits. Cbs-/-, but not Cbs+/- mice had increased expression of NR2B subunit, which is known to be relatively insensitive to homocysteine. Conclusions These data provide experimental evidence that even a mild increase in plasma total homocysteine can exacerbate cerebrovascular injury and suggest that N-methyl-D-aspartate receptor antagonism may represent a strategy to prevent reperfusion injury after acute ischemic stroke in patients with mild hyperhomocysteinemia.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Homocisteína/sangre , Hiperhomocisteinemia/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/prevención & control , Memantina/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Cistationina betasintasa/deficiencia , Cistationina betasintasa/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Homocistinuria/enzimología , Homocistinuria/genética , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/enzimología , Hiperhomocisteinemia/genética , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Receptores de N-Metil-D-Aspartato/metabolismo , Índice de Severidad de la Enfermedad
6.
Mutat Res ; 819-820: 111687, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31968288

RESUMEN

Methylene tetrahydrofolate reductase (MTHFR) is a flavoprotein, involved in one-carbon pathway and is responsible for folate and homocysteine metabolism. Regulation of MTHFR is pivotal for maintaining the cellular concentrations of methionine and SAM (S-adenosyl methionine) which are essential for the synthesis of nucleotides and amino acids, respectively. Therefore, mutations in MTHFR leads to its dysfunction resulting in conditions like homocystinuria, cardiovascular diseases, and neural tube defects in infants. Among these conditions, homocystinuria has been highly explored, as it manifests ocular disorders, cognitive disorders and skeletal abnormalities. Hence, in this study, we intend to explore the mutational landscape of human MTHFR isoform-1 (h.MTHFR-1) to decipher the most pathogenic variants pertaining to homocystinuria. Thus, a multilevel stringent prioritization of non-synonymous mutations in h.MTHFR-1 by integrative machine learning approaches was implemented to delineate highly deleterious variants based on its pathogenicity, impact on structural stability and functionality. Subsequently, extended molecular dynamics simulations and molecular docking studies were also integrated in order to prioritize the mutations that perturbs structural stability and functionality of h.MTHFR-1. In addition, displacement of Loop (Arg157-Tyr174) and helix α9 (His263-Ser272) involved in open/closed conformation of substrate binding domain were also probed to confirm the functional loss. On juxtaposed analysis, it was inferred that among 126 missense mutations screened, along with known pathogenic mutations (H127 T, A222 V, T227 M, F257 V and G387D) predicted that W500C, P254S and D585 N variants could be potentially driving homocystinuria. Thus, uncovering the prospects for inclusion of these mutations in diagnostic panels based on further experimental validations.


Asunto(s)
Homocistinuria/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/química , Mutación Missense , S-Adenosilmetionina/química , Tetrahidrofolatos/química , Sitio Alostérico , Secuencias de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Expresión Génica , Homocistinuria/enzimología , Homocistinuria/patología , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Aprendizaje Automático , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , NADP/química , NADP/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , S-Adenosilmetionina/metabolismo , Especificidad por Sustrato , Tetrahidrofolatos/metabolismo , Termodinámica
7.
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
8.
Mol Genet Metab ; 120(4): 325-336, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28291718

RESUMEN

Classical homocystinuria (HCU) due to inactivating mutation of cystathionine ß-synthase (CBS) is a poorly understood life-threatening inborn error of sulfur metabolism. A previously described cbs-/- mouse model exhibits a semi-lethal phenotype due to neonatal liver failure. The transgenic HO mouse model of HCU exhibits only mild liver injury and recapitulates multiple aspects of the disease as it occurs in humans. Disruption of the methionine cycle in HCU has the potential to impact multiple aspect of phospholipid (PL) metabolism by disruption of both the Kennedy pathway and phosphatidylethanolamine N-methyltransferase (PEMT) mediated synthesis of phosphatidylcholine (PC). Comparative metabolomic analysis of HO mouse liver revealed decreased levels of choline, and choline phosphate indicating disruption of the Kennedy pathway. Alterations in the relative levels of multiple species of PL included significant increases in PL degradation products consistent with enhanced membrane PL turnover. A significant decrease in PC containing 20:4n6 which primarily formed by the methylation of phosphatidylethanolamine to PC was consistent with decreased flux through PEMT. Hepatic expression of PEMT in both the cbs-/- and HO models is post-translationally repressed with decreased levels of PEMT protein and activity that inversely-correlates with the scale of liver injury. Failure to induce further repression of PEMT in HO mice by increased homocysteine, methionine and S-adenosylhomocysteine or depletion of glutathione combined with examination of multiple homocysteine-independent models of liver injury indicated that repression of PEMT in HCU is a consequence rather than a cause of liver injury. Collectively, our data show significant alteration of a broad range of hepatic PL and choline metabolism in HCU with the potential to contribute to multiple aspects of pathogenesis in this disease.


Asunto(s)
Colina/metabolismo , Homocistinuria/enzimología , Hígado/química , Fosfatidiletanolamina N-Metiltransferasa/metabolismo , Fosfolípidos/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Hígado/lesiones , Metabolómica , Ratones , Ratones Noqueados , Fosfatidiletanolamina N-Metiltransferasa/genética , Procesamiento Proteico-Postraduccional
9.
J Clin Invest ; 126(6): 2043-4, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27183384

RESUMEN

Inherited metabolic disorders are often characterized by the lack of an essential enzyme and are currently treated by dietary restriction and other strategies to replace the substrates or products of the missing enzyme. Patients with homocystinuria lack the enzyme cystathionine ß-synthase (CBS), and many of these individuals do not respond to current treatment protocols. In this issue of the JCI, Bublil and colleagues demonstrate that enzyme replacement therapy (ERT) provides long-term amelioration of homocystinuria-associated phenotypes in CBS-deficient murine models. A PEGylated form of CBS provided long-term stability and, when used in conjunction with the methylation agent betaine, dramatically increased survival in mice fed a normal diet. The results of this study provide one of the first examples of ERT for a metabolic disorder and suggest that PEGylated CBS should be further explored for use in patients.


Asunto(s)
Modelos Animales de Enfermedad , Homocistinuria/enzimología , Animales , Betaína , Cistationina betasintasa/deficiencia , Humanos , Ratones , Fenotipo
10.
Clin Chim Acta ; 458: 55-62, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27059523

RESUMEN

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


Asunto(s)
Betaína/uso terapéutico , Homocistinuria/tratamiento farmacológico , Vitamina B 12/uso terapéutico , Vitamina B 6/uso terapéutico , Betaína/administración & dosificación , Cistationina betasintasa/deficiencia , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Homocistinuria/enzimología , Humanos , Concentración de Iones de Hidrógeno , Estrés Oxidativo/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Vitamina B 12/administración & dosificación , Vitamina B 6/administración & dosificación
11.
Biochimie ; 126: 6-13, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26805382

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/metabolismo , Homocistinuria/tratamiento farmacológico , Homocistinuria/enzimología , S-Adenosilmetionina/análogos & derivados , S-Adenosilmetionina/farmacología , Estabilidad de Enzimas/efectos de los fármacos , Humanos , Cinética , S-Adenosilmetionina/química
12.
J Inherit Metab Dis ; 39(1): 115-24, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26025547

RESUMEN

BACKGROUND: Severe methylenetetrahydrofolate reductase (MTHFR) deficiency is a rare inborn defect disturbing the remethylation of homocysteine to methionine (<200 reported cases). This retrospective study evaluates clinical, biochemical genetic and in vitro enzymatic data in a cohort of 33 patients. METHODS: Clinical, biochemical and treatment data was obtained from physicians by using a questionnaire. MTHFR activity was measured in primary fibroblasts; genomic DNA was extracted from cultured fibroblasts. RESULTS: Thirty-three patients (mean age at follow-up 11.4 years; four deceased; median age at first presentation 5 weeks; 17 females) were included. Patients with very low (<1.5%) mean control values of enzyme activity (n = 14) presented earlier and with a pattern of feeding problems, encephalopathy, muscular hypotonia, neurocognitive impairment, apnoea, hydrocephalus, microcephaly and epilepsy. Patients with higher (>1.7-34.8%) residual enzyme activity had mainly psychiatric symptoms, mental retardation, myelopathy, ataxia and spasticity. Treatment with various combinations of betaine, methionine, folate and cobalamin improved the biochemical and clinical phenotype. During the disease course, patients with very low enzyme activity showed a progression of feeding problems, neurological symptoms, mental retardation, and psychiatric disease while in patients with higher residual enzyme activity, myelopathy, ataxia and spasticity increased. All other symptoms remained stable or improved in both groups upon treatment as did brain imaging in some cases. No clear genotype-phenotype correlation was obvious. DISCUSSION: MTHFR deficiency is a severe disease primarily affecting the central nervous system. Age at presentation and clinical pattern are correlated with residual enzyme activity. Treatment alleviates biochemical abnormalities and clinical symptoms partially.


Asunto(s)
Homocistinuria/enzimología , Homocistinuria/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Espasticidad Muscular/enzimología , Espasticidad Muscular/genética , Ataxia/genética , Betaína/uso terapéutico , Niño , Femenino , Ácido Fólico/uso terapéutico , Estudios de Asociación Genética/métodos , Homocistinuria/tratamiento farmacológico , Humanos , Discapacidad Intelectual/genética , Masculino , Metionina/uso terapéutico , Espasticidad Muscular/tratamiento farmacológico , Mutación/genética , Fenotipo , Trastornos Psicóticos/tratamiento farmacológico , Trastornos Psicóticos/enzimología , Trastornos Psicóticos/genética , Estudios Retrospectivos , Enfermedades de la Médula Espinal/genética , Vitamina B 12/uso terapéutico
13.
Pediatr Int ; 57(5): 884-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25939784

RESUMEN

BACKGROUND: Classic homocystinuria due to cystathionine ß-synthase (CBS) deficiency is an autosomal recessive disorder of sulfur metabolism. Clinical manifestations include mental retardation, dislocation of the optic lens (ectopia lentis), skeletal abnormalities and a tendency to thromboembolic episodes. We present the first mutational analysis of CBS in a Filipino patient with classic homocystinuria. METHODS: Genomic DNA was extracted from peripheral blood collected from a diagnosed Filipino patient with classic homocystinuria. The entire coding region of CBS (17 exons) was amplified using polymerase chain reaction and bidirectionally sequenced using standard protocols. RESULTS: The patient was found to be compound heterozygous for two novel mutations, g.13995G>A [c.982G>A; p.D328K] and g.15860-15868dupGCAGGAGCT [c.1083-1091dupGCAGGAGCT; p. Q362-L364dupQEL]. Four known single-nucleotide polymorphisms (rs234706, rs1801181, rs706208 and rs706209) were also detected in the present patient's CBS. The patient was heterozygous for all the identified alleles. CONCLUSIONS: This is the first mutational analysis of CBS done in a Filipino patient with classic homocystinuria who presented with a novel duplication mutation and a novel missense mutation. Homocystinuria due to CBS deficiency is a heterogeneous disorder at the molecular level.


Asunto(s)
Cistationina betasintasa/genética , ADN/genética , Homocistinuria/genética , Mutación , Adolescente , Alelos , Cistationina betasintasa/metabolismo , Análisis Mutacional de ADN , Femenino , Genotipo , Homocistinuria/enzimología , Humanos , Filipinas
14.
Nucleic Acids Res ; 43(9): 4627-39, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25878036

RESUMEN

The prevalent c.903+469T>C mutation in MTRR causes the cblE type of homocystinuria by strengthening an SRSF1 binding site in an ESE leading to activation of a pseudoexon. We hypothesized that other splicing regulatory elements (SREs) are also critical for MTRR pseudoexon inclusion. We demonstrate that the MTRR pseudoexon is on the verge of being recognized and is therefore vulnerable to several point mutations that disrupt a fine-tuned balance between the different SREs. Normally, pseudoexon inclusion is suppressed by a hnRNP A1 binding exonic splicing silencer (ESS). When the c.903+469T>C mutation is present two ESEs abrogate the activity of the ESS and promote pseudoexon inclusion. Blocking the 3'splice site or the ESEs by SSOs is effective in restoring normal splicing of minigenes and endogenous MTRR transcripts in patient cells. By employing an SSO complementary to both ESEs, we were able to rescue MTRR enzymatic activity in patient cells to approximately 50% of that in controls. We show that several point mutations, individually, can activate a pseudoexon, illustrating that this mechanism can occur more frequently than previously expected. Moreover, we demonstrate that SSO blocking of critical ESEs is a promising strategy to treat the increasing number of activated pseudoexons.


Asunto(s)
Anemia Megaloblástica/genética , Exones , Ferredoxina-NADP Reductasa/genética , Homocistinuria/genética , Mutación , Oligonucleótidos , Empalme del ARN , Secuencias Reguladoras de Ácido Ribonucleico , Anemia Megaloblástica/enzimología , Línea Celular , Células Cultivadas , Ferredoxina-NADP Reductasa/metabolismo , Células HEK293 , Homocistinuria/enzimología , Humanos , Sitios de Empalme de ARN
15.
J Inherit Metab Dis ; 38(2): 287-94, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25331909

RESUMEN

Classical homocystinuria is caused by mutations in the cystathionine ß-synthase (CBS) gene. Previous experiments in bacterial and yeast cells showed that many mutant CBS enzymes misfold and that chemical chaperones enable proper folding of a number of mutations. In the present study, we tested the extent of misfolding of 27 CBS mutations previously tested in E. coli under the more folding-permissive conditions of mammalian CHO-K1 cells and the ability of chaperones to rescue the conformation of these mutations. Expression of mutations in mammalian cells increased the median activity 16-fold and the amount of tetramers 3.2-fold compared with expression in bacteria. Subsequently, we tested the responses of seven selected mutations to three compounds with chaperone-like activity. Aminooxyacetic acid and 4-phenylbutyric acid exhibited only a weak effect. In contrast, heme arginate substantially increased the formation of mutant CBS protein tetramers (up to sixfold) and rescued catalytic activity (up to ninefold) of five out of seven mutations (p.A114V, p.K102N, p.R125Q, p.R266K, and p.R369C). The greatest effect of heme arginate was observed for the mutation p.R125Q, which is non-responsive to in vivo treatment with vitamin B(6). Moreover, the heme responsiveness of the p.R125Q mutation was confirmed in fibroblasts derived from a patient homozygous for this genetic variant. Based on these data, we propose that a distinct group of heme-responsive CBS mutations may exist and that the heme pocket of CBS may become an important target for designing novel therapies for homocystinuria.


Asunto(s)
Arginina/farmacología , Cistationina betasintasa/genética , Fibroblastos/efectos de los fármacos , Hemo/farmacología , Homocistinuria/tratamiento farmacológico , Chaperonas Moleculares/farmacología , Mutación , Deficiencias en la Proteostasis/tratamiento farmacológico , Animales , Células CHO , Dominio Catalítico , Cricetulus , Cistationina betasintasa/metabolismo , Femenino , Fibroblastos/enzimología , Predisposición Genética a la Enfermedad , Homocistinuria/diagnóstico , Homocistinuria/enzimología , Homocistinuria/genética , Homocigoto , Humanos , Modelos Moleculares , Fenotipo , Conformación Proteica , Pliegue de Proteína , Deficiencias en la Proteostasis/diagnóstico , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/genética , Relación Estructura-Actividad , Especificidad por Sustrato , Transfección
16.
Hum Mutat ; 35(10): 1195-202, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25044645

RESUMEN

Cystathionine beta-synthase (CBS) catalyzes the formation of cystathionine from homocysteine and serine. CBS is allosterically activated by S-adenosylmethionine (SAM), which binds to its C-terminal regulatory domain. Mutations in this domain lead to variants with high residual activity but lacking SAM activation. We characterized six C-terminal CBS variants (p.P427L, p.D444N, p.V449G, p.S500L, p.K523Sfs*18, and p.L540Q). To understand the effect of C-terminal mutations on the functional/structural properties of CBS, we performed dynamic light scattering, differential scanning fluorimetry, limited proteolysis, enzymatic characterization, and determination of SAM-binding affinity. Kinetic data confirm that the enzymatic function of these variants is not impaired. Although lacking SAM activation, the p.P427L and p.S500L were able to bind SAM at a lower extent than the wild type (WT), confirming that SAM binding and activation can be two independent events. At the structural level, the C-terminal variants presented various effects, either showing catalytic core instability and increased susceptibility toward aggregation or presenting with similar or higher stability than the WT. Our study highlights as the common feature to the C-terminal variants an impaired binding of SAM and no increase in enzymatic activity with physiological concentrations of the activator, suggesting the loss of regulation by SAM as a potential pathogenic mechanism.


Asunto(s)
Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Homocistinuria/enzimología , Mutación , Sitio Alostérico , Secuencia de Aminoácidos , Dominio Catalítico , Cistationina betasintasa/química , Homocistinuria/genética , Humanos , Cinética , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , S-Adenosilmetionina/metabolismo
18.
Gene ; 539(2): 270-4, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24534463

RESUMEN

High blood levels of homocysteine (Hcy) are found in patients affected by homocystinuria, a genetic disorder caused by deficiency of cystathionine ß-synthase (CBS) activity, as well as in nutritional deficiencies (vitamin B12 or folate) and in abnormal renal function. We previously demonstrated that lipid and protein oxidative damage is increased and the antioxidant defenses diminished in plasma of CBS-deficient patients, indicating that oxidative stress is involved in the pathophysiology of this disease. In the present work, we extended these investigations by evaluating DNA damage through the comet assay in peripheral leukocytes from CBS-deficient patients, as well as by analyzing of the in vitro effect of Hcy on DNA damage in white blood cells. We verified that DNA damage was significantly higher in the CBS-deficient patients under treatment based on a protein-restricted diet and pyridoxine, folic acid, betaine and vitamin B12 supplementation, when compared to controls. Furthermore, the in vitro study showed a concentration-dependent effect of Hcy inducing DNA damage. Taken together, the present data indicate that DNA damage occurs in treated CBS-deficient patients, possibly due to high Hcy levels.


Asunto(s)
Cistationina betasintasa/deficiencia , Cistationina betasintasa/genética , Daño del ADN , Homocisteína/sangre , Homocistinuria/genética , Adolescente , Adulto , Estudios de Casos y Controles , Niño , Ensayo Cometa , Cistationina betasintasa/sangre , Femenino , Estudios de Seguimiento , Homocistinuria/sangre , Homocistinuria/enzimología , Humanos , Masculino , Pronóstico , Adulto Joven
19.
Gene ; 534(2): 197-203, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-24211323

RESUMEN

Classical homocystinuria is the most commonly inherited disorder of sulfur metabolism, caused by the genetic alterations in human cystathionine beta-synthase (CBS) gene. In this study, we present comprehensive clinical findings and the genetic basis of homocystinuria in a cohort of Turkish patients. Excluding some CBS mutations, detailed genotype-phenotype correlation for different CBS mutations has not been established in literature. We aimed to make clinical subgroups according to main clinical symptoms and discussed these data together with mutational analysis results from our patients. Totally, 16 different mutations were identified; twelve of which had already been reported, and four are novel (p.N93Y, p.L251P, p.D281V and c.829-2A>T). The probands were classified into three major groups according to the clinical symptoms caused by these mutations. A psychomotor delay was the most common diagnostic symptom (n=12, 46.2% neurological presentation), followed by thromboembolic events (n=6, 23.1% vascular presentation) and lens ectopia, myopia or marfanoid features (n=5, 19.2% connective tissue presentation). Pyridoxine responsiveness was 7.7%; however, with partial responsive probands, the ratio was 53.9%. In addition, five thrombophilic nucleotide changes including MTHFR c.677 C>T and c.1298 A>C, Factor V c.1691 G>A, Factor II c.20210 G>A, and SERPINE1 4G/5G were investigated to assess their contributions to the clinical spectrum. We suggest that the effect of these polymorphisms on clinical phenotype of CBS is not very clear since the distribution of thrombophilic polymorphisms does not differ among specific groups. This study provides molecular findings of 26 Turkish probands with homocystinuria and discusses the clinical presentations and putative effects of the CBS mutations.


Asunto(s)
Cistationina betasintasa/genética , Homocistinuria/diagnóstico , Trombosis de los Senos Intracraneales/diagnóstico , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN/métodos , Femenino , Estudios de Asociación Genética/métodos , Predisposición Genética a la Enfermedad , Genotipo , Homocistinuria/enzimología , Homocistinuria/epidemiología , Homocistinuria/genética , Humanos , Lactante , Masculino , Mutación , Fenotipo , Polimorfismo Genético , Prevalencia , Trombosis de los Senos Intracraneales/enzimología , Trombosis de los Senos Intracraneales/epidemiología , Trombosis de los Senos Intracraneales/genética , Turquía/epidemiología , Adulto Joven
20.
J Inherit Metab Dis ; 37(2): 245-54, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23974653

RESUMEN

A reduced response of cystathionine beta-synthase (CBS) to its allosteric activator S-adenosylmethionine (SAM) has been reported to be a cause of CBS dysfunction in homocystinuria patients. In this work we performed a retrospective analysis of fibroblast data from 62 homocystinuria patients and found that 13 of them presented a disturbed SAM activation. Their genotypic background was identified and the corresponding CBS mutant proteins were produced in E. coli. Nine distinct mutations were detected in 22 independent alleles: the novel mutations p.K269del, p.P427L, p.S500L and p.L540Q; and the previously described mutations p.P49L, p.C165Rfs*2, p.I278T, p.R336H and p.D444N. Expression levels and residual enzyme activities, determined in the soluble fraction of E. coli lysates, strongly correlated with the localization of the affected amino acid residue. C-terminal mutations lead to activities in the range of the wild-type CBS and to oligomeric forms migrating faster than tetramers, suggesting an abnormal conformation that might be responsible for the lack of SAM activation. Mutations in the catalytic core were associated with low protein expression levels, decreased enzyme activities and a higher content of high molecular mass forms. Furthermore, the absence of SAM activation found in the patients' fibroblasts was confirmed for all but one of the characterized recombinant proteins (p.P49L). Our study experimentally supports a deficient regulation of CBS by SAM as a frequently found mechanism in CBS deficiency, which should be considered not only as a valuable diagnostic tool but also as a potential target for the development of new therapeutic approaches in classical homocystinuria.


Asunto(s)
Cistationina betasintasa/genética , Homocistinuria/enzimología , Homocistinuria/genética , Mutación , S-Adenosilmetionina/genética , Alelos , Células Cultivadas , Cistationina betasintasa/metabolismo , Escherichia coli/genética , Fibroblastos/enzimología , Fibroblastos/metabolismo , Fibroblastos/patología , Genotipo , Homocistinuria/metabolismo , Homocistinuria/patología , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estudios Retrospectivos , S-Adenosilmetionina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...