Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 240
Filtrar
1.
Mol Med ; 30(1): 3, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172668

RESUMEN

BACKGROUND: Lesch-Nyhan disease (LND) is a severe neurological disorder caused by the genetic deficiency of hypoxanthine-guanine phosphoribosyltransferase (HGprt), an enzyme involved in the salvage synthesis of purines. To compensate this deficiency, there is an acceleration of the de novo purine biosynthetic pathway. Most studies have failed to find any consistent abnormalities of purine nucleotides in cultured cells obtained from the patients. Recently, it has been shown that 5-aminoimidazole-4-carboxamide riboside 5'-monophosphate (ZMP), an intermediate of the de novo pathway, accumulates in LND fibroblasts maintained with RPMI containing physiological levels (25 nM) of folic acid (FA), which strongly differs from FA levels of regular cell culture media (2200 nM). However, RPMI and other standard media contain non-physiological levels of many nutrients, having a great impact in cell metabolism that does not precisely recapitulate the in vivo behavior of cells. METHODS: We prepared a new culture medium containing physiological levels of all nutrients, including vitamins (Plasmax-PV), to study the potential alterations of LND fibroblasts that may have been masked by the usage of non-physiological media. We quantified ZMP accumulation under different culture conditions and evaluated the activity of two known ZMP-target proteins (AMPK and ADSL), the mRNA expression of the folate carrier SLC19A1, possible mitochondrial alterations and functional consequences in LND fibroblasts. RESULTS: LND fibroblasts maintained with Plasmax-PV show metabolic adaptations such a higher glycolytic capacity, increased expression of the folate carrier SCL19A1, and functional alterations such a decreased mitochondrial potential and reduced cell migration compared to controls. These alterations can be reverted with high levels of folic acid, suggesting that folic acid supplements might be a potential treatment for LND. CONCLUSIONS: A complete physiological cell culture medium reveals new alterations in Lesch-Nyhan disease. This work emphasizes the importance of using physiological cell culture conditions when studying a metabolic disorder.


Asunto(s)
Síndrome de Lesch-Nyhan , Humanos , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/metabolismo , Hipoxantina Fosforribosiltransferasa/genética , Hipoxantina Fosforribosiltransferasa/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Ácido Fólico
2.
J Biochem Mol Toxicol ; 38(1): e23558, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37865952

RESUMEN

This study explored the effect of naringin (NAR) on HGPRT1 deficiency and hyperuricemia through NOS-cAMP-PKA and BDNF/TrkB signaling pathways induced by caffeine (CAF) and KBrO3 in a rat model. Sixty-three adult male albino rats were randomly assigned into nine (n = 7) groups. Group I: control animals, Group II was treated with 100 mg/kg KBrO3 , Group III was treated with 250 mg/kg CAF, Group IV was treated with 100 mg/kg KBrO3 + 250 mg/kg CAF, Group V was administered with 100 mg/kg KBrO3 + 100 mg/kg haloperidol, Group VI was administered with 100 mg/kg KBrO3 + 50 mg/kg NAR, Group VII was administered with 500 mg/kg CAF + 50 mg/kg NAR, and Group VIII was administered with 100 mg/kg KBrO3 + 250 mg/kg CAF + 50 mg/kg NAR. Finally, group IX was treated with 50 mg/kg NAR. The exposure of rats to KBrO3 and CAF for 21 days induced renal dysfunction linked with Lesch-Nyhan disease. NAR obliterated renal dysfunction linked with Lesch-Nyhan disease by decreasing uric acid, renal malondialdehyde level, inhibiting the activities of arginase, and phosphodiesterase-51 (PDE-51) with corresponding upregulation of brain derived-neurotrophic factor and its receptor (BDNF-TrkB), Bcl11b, HGPRT1, and DARPP-32. Additionally, renal failure related to Lesch-Nyhan disease was remarkably corrected by NAR as shown by the reduced activities of AChE and enzymes of ATP hydrolysis (ATPase, AMPase, and ADA) with affiliated increase in the NO level. This study therefore validates NAR as nontoxic and effective chemotherapy against kidney-related Lesch-Nyhan disease by mitigating effects of toxic food additives and enzymes of ATP-hydrolysis via NOS-cAMP-PKA and BDNF/TrkB signaling pathways.


Asunto(s)
Flavanonas , Síndrome de Lesch-Nyhan , Insuficiencia Renal , Masculino , Ratas , Animales , Síndrome de Lesch-Nyhan/tratamiento farmacológico , Síndrome de Lesch-Nyhan/metabolismo , Factor Neurotrófico Derivado del Encéfalo , Cafeína , Adenosina Trifosfato
3.
Cell Mol Life Sci ; 79(6): 341, 2022 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-35660973

RESUMEN

In Lesch-Nyhan disease (LND), deficiency of the purine salvage enzyme hypoxanthine guanine phosphoribosyl transferase (HGprt) leads to a characteristic neurobehavioral phenotype dominated by dystonia, cognitive deficits and incapacitating self-injurious behavior. It has been known for decades that LND is associated with dysfunction of midbrain dopamine neurons, without overt structural brain abnormalities. Emerging post mortem and in vitro evidence supports the hypothesis that the dopaminergic dysfunction in LND is of developmental origin, but specific pathogenic mechanisms have not been revealed. In the current study, HGprt deficiency causes specific neurodevelopmental abnormalities in mice during embryogenesis, particularly affecting proliferation and migration of developing midbrain dopamine (mDA) neurons. In mutant embryos at E14.5, proliferation was increased, accompanied by a decrease in cell cycle exit and the distribution and orientation of dividing cells suggested a premature deviation from their migratory route. An abnormally structured radial glia-like scaffold supporting this mDA neuronal migration might lie at the basis of these abnormalities. Consequently, these abnormalities were associated with an increase in area occupied by TH+ cells and an abnormal mDA subpopulation organization at E18.5. Finally, dopaminergic innervation was disorganized in prefrontal and decreased in HGprt deficient primary motor and somatosensory cortices. These data provide direct in vivo evidence for a neurodevelopmental nature of the brain disorder in LND. Future studies should not only focus the specific molecular mechanisms underlying the reported neurodevelopmental abnormalities, but also on optimal timing of therapeutic interventions to rescue the DA neuron defects, which may also be relevant for other neurodevelopmental disorders.


Asunto(s)
Síndrome de Lesch-Nyhan , Animales , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Hipoxantina Fosforribosiltransferasa/genética , Hipoxantina Fosforribosiltransferasa/metabolismo , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/metabolismo , Mesencéfalo/metabolismo , Ratones
4.
Proc Natl Acad Sci U S A ; 117(22): 12071-12079, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32430324

RESUMEN

Lesch-Nyhan disease (LND), caused by a deficient salvage purine pathway, is characterized by severe neurological manifestations and uric acid overproduction. However, uric acid is not responsible for brain dysfunction, and it has been suggested that purine nucleotide depletion, or accumulation of other toxic purine intermediates, could be more relevant. Here we show that purine alterations in LND fibroblasts depend on the level of folic acid in the culture media. Thus, physiological levels of folic acid induce accumulation of 5-aminoimidazole-4-carboxamide riboside 5'-monophosphate (ZMP), an intermediary of de novo purine biosynthetic pathway, and depletion of ATP. Additionally, Z-nucleotide derivatives (AICAr, AICA) are detected at high levels in the urine of patients with LND and its variants (hypoxanthine-guanine phosphoribosyltransferase [HGprt]-related neurological dysfunction and HGprt-related hyperuricemia), and the ratio of AICAr/AICA is significantly increased in patients with neurological problems (LND and HGprt-related neurological dysfunction). Moreover, AICAr is present in the cerebrospinal fluid of patients with LND, but not in control individuals. We hypothesize that purine alterations detected in LND fibroblasts may also occur in the brain of patients with LND.


Asunto(s)
Ácido Fólico/análisis , Síndrome de Lesch-Nyhan/etiología , Purinas/metabolismo , Adenosina Trifosfato/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/metabolismo , Técnicas de Cultivo de Célula , Medios de Cultivo Condicionados/química , Fibroblastos/metabolismo , Humanos , Hipoxantina Fosforribosiltransferasa/metabolismo , Síndrome de Lesch-Nyhan/metabolismo , Ribonucleótidos/metabolismo
5.
J Neurogenet ; 36(2-3): 81-87, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36226509

RESUMEN

Lesch-Nyhan disease (LND) is a neurodevelopmental disorder caused by variants in the HPRT1 gene, which encodes the enzyme hypoxanthine-guanine phosphoribosyl transferase (HGprt). HGprt deficiency provokes numerous metabolic changes which vary among different cell types, making it unclear which changes are most relevant for abnormal neural development. To begin to elucidate the consequences of HGprt deficiency for developing human neurons, neural stem cells (NSCs) were prepared from 6 induced pluripotent stem cell (iPSC) lines from individuals with LND and compared to 6 normal healthy controls. For all 12 lines, gene expression profiles were determined by RNA-seq and protein expression profiles were determined by shotgun proteomics. The LND lines revealed significant changes in expression of multiple genes and proteins. There was little overlap in findings between iPSCs and NSCs, confirming the impact of HGprt deficiency depends on cell type. For NSCs, gene expression studies pointed towards abnormalities in WNT signaling, which is known to play a role in neural development. Protein expression studies pointed to abnormalities in the mitochondrial F0F1 ATPase, which plays a role in maintaining cellular energy. These studies point to some mechanisms that may be responsible for abnormal neural development in LND.


Asunto(s)
Síndrome de Lesch-Nyhan , Células-Madre Neurales , Humanos , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/metabolismo , Hipoxantina Fosforribosiltransferasa/genética , Hipoxantina Fosforribosiltransferasa/metabolismo , Guanina/metabolismo , Adenosina Trifosfatasas , Hipoxantinas
6.
Mol Biol Rep ; 48(1): 701-708, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33393008

RESUMEN

Elongator is a multi-subunit protein complex bearing six different protein subunits, Elp1 to -6, that are highly conserved among eukaryotes. Elp2 is the second major subunit of Elongator and, together with Elp1 and Elp3, form the catalytic core of this essential complex. Pathogenic variants that affect the structure and function of the Elongator complex may cause neurodevelopmental disorders. Here, we report on a new family with three children affected with a severe form of intellectual disability along with spastic tetraparesis, choreoathetosis, and self injury. Molecular genetic analyses reveal a homozygous missense variant in the ELP2 gene (NM_018255.4 (ELP2): c.1385G > A (p.Arg462Gln)), while in silico studies suggest a loss of electrostatic interactions that may contribute to the overall stability of the encoded protein. We also include a comparison of the patients with ELP2-related neurodevelopmental disorder to those previously reported in the literature. Apart from being affected with intellectual disability, we have extremely limited clinical knowledge about patients harboring ELP2 variants. Besides providing support to the causal role of p.Arg462Gln in ELP2-related neurodevelopmental disorder, we add self-injurious behavior to the clinical phenotypic repertoire of the disease.


Asunto(s)
Discapacidad Intelectual/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Síndrome de Lesch-Nyhan/genética , Paresia/genética , Polimorfismo de Nucleótido Simple , Adolescente , Secuencia de Aminoácidos , Consanguinidad , Familia , Femenino , Expresión Génica , Homocigoto , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Síndrome de Lesch-Nyhan/diagnóstico , Síndrome de Lesch-Nyhan/metabolismo , Síndrome de Lesch-Nyhan/patología , Masculino , Persona de Mediana Edad , Modelos Moleculares , Paresia/diagnóstico , Paresia/metabolismo , Paresia/patología , Linaje , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Electricidad Estática , Turquía , Secuenciación del Exoma , Adulto Joven
7.
J Inherit Metab Dis ; 42(1): 178-185, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30740729

RESUMEN

Lesch-Nyhan disease (LND) is a rare X-linked genetic disorder, with complete hypoxanthine-guanine phosphoribosyltransferase (HGPRT) deficiency, uric acid (UA), hypoxanthine and xanthine accumulation, and a devastating neurologic syndrome. UA excess, causing renal failure, is commonly decreased by xanthine oxidoreductase (XOR) inhibitors, such as allopurinol, yielding a xanthine and hypoxanthine increase. Xanthine accumulation may result in renal stones, while hypoxanthine excess seems involved in the neurological disorder. Inhibition of purine nucleoside phosphorylase (PNP) represents a different strategy for lowering urate. PNP catalyzes the cleavage of purine ribo- and d-ribo-nucleosides into ribose/deoxyribose phosphate and free bases, starting catabolism to uric acid. Clinical trials demonstrated that PNP inhibitors, initially developed as anticancer drugs, lowered UA in some gouty patients, in association or not with allopurinol. The present study tested the reliability of an analogue of immucillin-G (C1a), a PNP inhibitor, as a therapy for urate, hypoxanthine, and xanthine excess in LND patients by blocking hypoxanthine production upstream. The therapeutic aim is to limit the administration of XOR inhibitors to LND patients by supplying the PNP inhibitor in low doses, avoiding d-nucleoside toxicity. We report studies conducted in primary cultures of skin fibroblasts from controls and LND patients grown in the presence of the PNP inhibitor. Cell viability, oxypurine release in culture medium, and endocellular nucleotide pattern have been monitored in different growth conditions (inhibitor concentration, time, added inosine). Our results demonstrate effective PNP inhibition by low inhibitor concentration, with reduced hypoxanthine release, and no appreciable toxicity in control or patient cells, suggesting a new therapeutic strategy for LND hyperuricemia.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hiperuricemia/tratamiento farmacológico , Síndrome de Lesch-Nyhan/tratamiento farmacológico , Purina-Nucleósido Fosforilasa/antagonistas & inhibidores , Pirimidinonas/farmacología , Pirroles/farmacología , Alopurinol/farmacología , Células Cultivadas , Humanos , Hiperuricemia/metabolismo , Hipoxantina/farmacología , Hipoxantina Fosforribosiltransferasa/metabolismo , Síndrome de Lesch-Nyhan/metabolismo , Purinas/metabolismo , Reproducibilidad de los Resultados , Ácido Úrico/metabolismo , Xantina/farmacología
8.
Metab Brain Dis ; 34(5): 1335-1340, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31129767

RESUMEN

Lesch-Nyhan syndrome (LNS) is an X-linked recessive disorder caused by mutations in the HPRT1 gene. The clinical features and mutation spectrum of 26 Korean LNS patients from 23 unrelated families were retrospectively reviewed. The HPRT1 gene was analyzed by direct sequencing of genomic DNA. The median age at diagnosis was 2.3 years (range, 4 months-22.6 years) and the initial presenting features included developmental delay, orange colored urine, and self-injurious behaviors. Most patients were wheelchair-bound and suffered from urinary complications and neurologic problems such as self-mutilation and developmental delay. Twenty different mutations in HPRT1 were identified among 23 independent pedigrees, including six novel mutations. The most common mutation type was truncating mutations including nonsense and frameshift mutations (45%). Large deletions in the HPRT1 gene were identified in exon 1, exons 5-6, exons 1-9, and at chr X:134,459,540-134,467,241 (7702 bp) including the 5'-untranslated region, exon 1, and a portion of intron 1. In conclusion, this study describes the phenotypic spectrum of LNS and has identified 20 mutations from 23 Korean families, including six novel mutations in Korean patients with LNS.


Asunto(s)
Exones , Hipoxantina Fosforribosiltransferasa/genética , Síndrome de Lesch-Nyhan/diagnóstico , Mutación , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Humanos , Hipoxantina Fosforribosiltransferasa/metabolismo , Lactante , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/metabolismo , Masculino , Linaje , República de Corea , Estudios Retrospectivos , Adulto Joven
9.
Mol Genet Metab ; 118(3): 145-146, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27216368

RESUMEN

Lesch-Nyhan disease (LND) is a rare X-chromosomal purine metabolism disorder. LND is characterized by self-injurious behavior (SIB) for which there is no drug treatment. This commentary places a recent clinical study by Khasnavis et al. (Mol. Genetic. Metab., in press) on drug treatment of SIB into a broader context. Although the study by Khasnavis et al. was no break-through in terms of "positive" results, nonetheless, it presents an excellent model of how clinical studies in general and clinical studies on rare diseases should be conducted.


Asunto(s)
Benzazepinas/efectos adversos , Antagonistas de Dopamina/efectos adversos , Síndrome de Lesch-Nyhan/tratamiento farmacológico , Benzazepinas/uso terapéutico , Ensayos Clínicos Fase II como Asunto , Estudios Cruzados , Antagonistas de Dopamina/uso terapéutico , Humanos , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/metabolismo , Receptores de Dopamina D2/metabolismo , Tamaño de la Muestra , Insuficiencia del Tratamiento
10.
Mol Genet Metab ; 117(4): 401-6, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26922636

RESUMEN

Lesch-Nyhan disease (LND) is an inherited metabolic disorder characterized by the overproduction of uric acid and distinct behavioral, cognitive, and motor abnormalities. The most challenging clinical problem is self-injurious behavior (SIB), which includes self-biting, self-hitting, self-abrasion, and other features. Currently, these behaviors are managed by behavioral extinction, sedatives, physical restraints, and removal of teeth. More effective treatments are needed. Pre-clinical studies have led to the hypothesis that D1-dopamine receptor antagonists may provide useful treatments for SIB in LND. Ecopipam is one such selective D1-dopamine receptor antagonist. This report summarizes results of a dose-escalation study of the safety and tolerability of ecopipam in 5 subjects with LND. The results suggest that ecopipam is well tolerated, with sedation being the most common dose-limiting event. Several exploratory measures also suggest ecopipam might reduce SIB in this population. These results support the hypothesis that D1-dopamine receptor antagonists may be useful for suppressing SIB in LND, and encourage further studies of efficacy.


Asunto(s)
Benzazepinas/uso terapéutico , Antagonistas de Dopamina/uso terapéutico , Síndrome de Lesch-Nyhan/tratamiento farmacológico , Receptores de Dopamina D1/antagonistas & inhibidores , Adolescente , Adulto , Benzazepinas/administración & dosificación , Benzazepinas/efectos adversos , Niño , Antagonistas de Dopamina/administración & dosificación , Antagonistas de Dopamina/efectos adversos , Humanos , Síndrome de Lesch-Nyhan/diagnóstico , Síndrome de Lesch-Nyhan/metabolismo , Masculino , Persona de Mediana Edad , Conducta Autodestructiva , Resultado del Tratamiento , Adulto Joven
11.
J Inherit Metab Dis ; 39(1): 85-91, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26067813

RESUMEN

Lesch-Nyhan disease (LND) is a rare, X-linked recessive neurodevelopmental disorder caused by deficiency of hypoxanthine-guanine phosphoribosyltransferase (HGprt), an enzyme in the purine salvage pathway. HGprt has two functions; it recycles hypoxanthine and guanine. Which of these two functions is more relevant for pathogenesis is unclear because some evidence points to hypoxanthine recycling, but other evidence points to guanine recycling. In this study, we selectively assayed hypoxanthine (Hprt) and guanine (Gprt) recycling in skin fibroblasts from 17 persons with LND, 11 with an attenuated variant of the disease (LNV), and 19 age-, sex-, and race-matched healthy controls (HC). Activity levels of both enzymes differed across groups (p < 0.0001), but only Gprt distinguished patients with LND from those with LNV (p < 0.05). Gprt also showed slightly stronger correlations than Hprt with 13 of 14 measures of the clinical phenotype, including the severity of dystonia, cognitive impairment, and behavioral abnormalities. These findings suggest that loss of guanine recycling might be more closely linked to the LND/LNV phenotype than loss of hypoxanthine recycling.


Asunto(s)
Guanina/metabolismo , Hipoxantina/metabolismo , Síndrome de Lesch-Nyhan/metabolismo , Adolescente , Adulto , Estudios de Casos y Controles , Fibroblastos/metabolismo , Humanos , Hipoxantina Fosforribosiltransferasa/deficiencia , Hipoxantina Fosforribosiltransferasa/metabolismo , Síndrome de Lesch-Nyhan/genética , Masculino , Fenotipo , Purinas/metabolismo , Piel/metabolismo , Adulto Joven
12.
Mol Genet Metab ; 114(1): 55-61, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25481104

RESUMEN

Mutations in the HPRT1 gene, which encodes the purine salvage enzyme hypoxanthine-guanine phosphoribosyltransferase (HGprt), cause Lesch-Nyhan disease (LND) and more mildly affected Lesch-Nyhan variants. Prior studies have suggested a strong correlation between residual hypoxanthine recycling activity and disease severity. However, the relevance of guanine recycling and compensatory changes in the de novo synthesis of purines has received little attention. In the current studies, fibroblast cultures were established for 21 healthy controls and 36 patients with a broad spectrum of disease severity related to HGprt deficiency. We assessed hypoxanthine recycling, guanine recycling, steady-state purine pools, and de novo purine synthesis. There was a strong correlation between disease severity and either hypoxanthine or guanine recycling. Intracellular purines were normal in the HGprt-deficient fibroblasts, but purine wasting was evident as increased purine metabolites excreted from the cells. The normal intracellular purines in the HGprt-deficient fibroblasts were likely due in part to a compensatory increase in purine synthesis, as demonstrated by a significant increase in purinosomes. However, the increase in purine synthesis did not appear to correlate with disease severity. These results refine our understanding of the potential sources of phenotypic heterogeneity in LND and its variants.


Asunto(s)
Guanina/metabolismo , Hipoxantina Fosforribosiltransferasa/deficiencia , Hipoxantina Fosforribosiltransferasa/metabolismo , Hipoxantina/metabolismo , Síndrome de Lesch-Nyhan/metabolismo , Purinas/biosíntesis , Células Cultivadas , Fibroblastos , Humanos , Purinas/metabolismo
13.
Mol Genet Metab ; 114(4): 570-579, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25769394

RESUMEN

The importance of specific pathways of purine metabolism for normal brain function is highlighted by several inherited disorders, such as Lesch-Nyhan disease (LND). In this disorder, deficiency of the purine recycling enzyme, hypoxanthine-guanine phosphoribosyltransferase (HGprt), causes severe neurological and behavioral abnormalities. Despite many years of research, the mechanisms linking the defect in purine recycling to the neurobehavioral abnormalities remain unclear. In the current studies, an unbiased approach to the identification of potential mechanisms was undertaken by examining changes in protein expression in a model of HGprt deficiency based on the dopaminergic rat PC6-3 line, before and after differentiation with nerve growth factor (NGF). Protein expression profiles of 5 mutant sublines carrying different mutations affecting HGprt enzyme activity were compared to the HGprt-competent parent line using the method of stable isotopic labeling by amino acids in cell culture (SILAC) followed by denaturing gel electrophoresis with liquid chromatography and tandem mass spectrometry (LC-MS/MS) of tryptic digests, and subsequent identification of affected biochemical pathways using the Database for Annotation, Visualization and Integrated Discovery (DAVID) functional annotation chart analysis. The results demonstrate that HGprt deficiency causes broad changes in protein expression that depend on whether the cells are differentiated or not. Several of the pathways identified reflect predictable consequences of defective purine recycling. Other pathways were not anticipated, disclosing previously unknown connections with purine metabolism and novel insights into the pathogenesis of LND.


Asunto(s)
Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/metabolismo , Proteoma , Purinas/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Dopamina/metabolismo , Neuronas Dopaminérgicas/fisiología , Hipoxantina Fosforribosiltransferasa/deficiencia , Hipoxantina Fosforribosiltransferasa/genética , Factor de Crecimiento Nervioso , Ratas , Espectrometría de Masas en Tándem , Transcriptoma
14.
Hum Mol Genet ; 21(3): 609-22, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22042773

RESUMEN

Mutations in the gene encoding the purine biosynthetic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT) cause the intractable neurodevelopmental Lesch-Nyhan disease (LND) associated with aberrant development of brain dopamine pathways. In the current study, we have identified an increased expression of the microRNA miR181a in HPRT-deficient human dopaminergic SH-SY5Y neuroblastoma cells. Among the genes potentially regulated by miR181a are several known to be required for neural development, including Engrailed1 (En1), Engrailed2 (En2), Lmx1a and Brn2. We demonstrate that these genes are down-regulated in HPRT-deficient SH-SY5Y cells and that over-expression of miR181a significantly reduces endogenous expression of these genes and inhibits translation of luciferase plasmids bearing the En1/2 or Lmx1a 3'UTR miRNA-binding elements. Conversely, inhibition of miR181a increases the expression of these genes and enhances translation of luciferase constructs bearing the En1/2 and Lmx1a 3'UTR miRNA-binding sequences. We also demonstrate that key neurodevelopmental genes (e.g. Nurr1, Pitx3, Wnt1 and Mash1) known to be functional partners of Lmx1a and Brn2 are also markedly down-regulated in SH-SY5Y cells over-expressing miR181a and in HPRT-deficient cells. Our findings in SH-SY5Y cells demonstrate that HPRT deficiency is accompanied by dysregulation of some of the important pathways that regulate the development of dopaminergic neurons and dopamine pathways and that this defect is associated with and possibly due at least partly to aberrant expression of miR181a. Because aberrant expression of miR181a is not as apparent in HPRT-deficient LND fibroblasts, the relevance of the SH-SY5Y neuroblastoma cells to human disease remains to be proven. Nevertheless, we propose that these pleiotropic neurodevelopment effects of miR181a may play a role in the pathogenesis of LND.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Regulación de la Expresión Génica , Hipoxantina Fosforribosiltransferasa/deficiencia , Síndrome de Lesch-Nyhan/genética , MicroARNs/metabolismo , Regiones no Traducidas 3' , Línea Celular Tumoral , Células Cultivadas , Regulación hacia Abajo , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Hipoxantina Fosforribosiltransferasa/antagonistas & inhibidores , Hipoxantina Fosforribosiltransferasa/genética , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Síndrome de Lesch-Nyhan/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores del Dominio POU/genética , Factores del Dominio POU/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Bioinformatics ; 29(24): 3191-8, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24078686

RESUMEN

MOTIVATION: Mathematical modeling and optimization have been used for detecting enzyme targets in human metabolic disorders. Such optimal drug design methods are generally differentiated as two stages, identification and decision-making, to find optimal targets. We developed a unified method named fuzzy equal metabolic adjustment to formulate an optimal enzyme target design problem for drug discovery. The optimization framework combines the identification of enzyme targets and a decision-making strategy simultaneously. The objectives of this algorithm include evaluations of the therapeutic effect of target enzymes, the adverse effects of drugs and the minimum effective dose (MED). RESULTS: An existing generalized mass action system model of human uric acid (UA) metabolism was used to formulate the fuzzy optimization method for detecting two types of enzymopathies: hyperuricemia caused by phosphoribosylpyrophosphate synthetase (PRPPS) overactivity and Lesch-Nyhan syndrome. The fuzzy objectives were set so that the concentrations of the metabolites were as close as possible to the healthy levels. The target design included a diet control of ribose-5-phospahate (R5P). The diet control of R5P served as an extra remedy to reduce phosphate uptake entering the purine metabolic pathway, so that we could obtain a more satisfactory treatment than obtained for those without a diet control. Moreover, enhancing UA excretion resulted in an effective treatment of hyperuricemia caused by PRPPS overactivity. This result correlates with using probenecid and benbromazone, which are uricosuric agents present in current clinical medications. By contrast, the Lesch-Nyhan syndrome required at least three enzyme targets to cure hyperuricemia.


Asunto(s)
Algoritmos , Diseño de Fármacos , Hiperuricemia/metabolismo , Síndrome de Lesch-Nyhan/metabolismo , Modelos Biológicos , Ácido Úrico/metabolismo , Dieta , Descubrimiento de Drogas , Lógica Difusa , Humanos , Hiperuricemia/etiología , Hiperuricemia/prevención & control , Cinética , Síndrome de Lesch-Nyhan/complicaciones , Síndrome de Lesch-Nyhan/etiología , Síndrome de Lesch-Nyhan/prevención & control , Redes y Vías Metabólicas , Probenecid/farmacología , Errores Innatos del Metabolismo de la Purina-Pirimidina/complicaciones , Purinas/metabolismo , Ribosa-Fosfato Pirofosfoquinasa/metabolismo , Ribosamonofosfatos/metabolismo , Uricosúricos/farmacología
17.
Elife ; 122024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38700995

RESUMEN

Adenine phosphoribosyltransferase (APRT) and hypoxanthine-guanine phosphoribosyltransferase (HGPRT) are two structurally related enzymes involved in purine recycling in humans. Inherited mutations that suppress HGPRT activity are associated with Lesch-Nyhan disease (LND), a rare X-linked metabolic and neurological disorder in children, characterized by hyperuricemia, dystonia, and compulsive self-injury. To date, no treatment is available for these neurological defects and no animal model recapitulates all symptoms of LND patients. Here, we studied LND-related mechanisms in the fruit fly. By combining enzymatic assays and phylogenetic analysis, we confirm that no HGPRT activity is expressed in Drosophila melanogaster, making the APRT homolog (Aprt) the only purine-recycling enzyme in this organism. Whereas APRT deficiency does not trigger neurological defects in humans, we observed that Drosophila Aprt mutants show both metabolic and neurobehavioral disturbances, including increased uric acid levels, locomotor impairments, sleep alterations, seizure-like behavior, reduced lifespan, and reduction of adenosine signaling and content. Locomotor defects could be rescued by Aprt re-expression in neurons and reproduced by knocking down Aprt selectively in the protocerebral anterior medial (PAM) dopaminergic neurons, the mushroom bodies, or glia subsets. Ingestion of allopurinol rescued uric acid levels in Aprt-deficient mutants but not neurological defects, as is the case in LND patients, while feeding adenosine or N6-methyladenosine (m6A) during development fully rescued the epileptic behavior. Intriguingly, pan-neuronal expression of an LND-associated mutant form of human HGPRT (I42T), but not the wild-type enzyme, resulted in early locomotor defects and seizure in flies, similar to Aprt deficiency. Overall, our results suggest that Drosophila could be used in different ways to better understand LND and seek a cure for this dramatic disease.


Asunto(s)
Drosophila melanogaster , Síndrome de Lesch-Nyhan , Animales , Drosophila melanogaster/fisiología , Drosophila melanogaster/genética , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/metabolismo , Purinas/metabolismo , Modelos Animales de Enfermedad , Conducta Animal , Hipoxantina Fosforribosiltransferasa/genética , Hipoxantina Fosforribosiltransferasa/metabolismo , Hipoxantina Fosforribosiltransferasa/deficiencia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Locomoción
18.
BMC Genomics ; 14: 773, 2013 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-24206569

RESUMEN

BACKGROUND: Transcription activator-like effector nucleases (TALENs) have emerged as a tool for enabling targeted gene editing and disruption in difficult systems, such as human pluripotent stem cells (hPSCs). The modular architecture of TAL effectors theoretically enables targeting of any genomic locus and several cloning systems for custom TALEN assembly have recently been established. However, there is a lack of versatile TALEN expression systems applicable to hPSCs. RESULTS: Here, we extend an existing TALE assembly system by a dual set of expression vectors for efficient application of TALEN technology in hPSCs. This is characterized by improved TALEN architecture as well as antibiotic resistance and fluorescent reporter cassettes, thus enabling enrichment for transfected cells. Improved functionality of the combined system was demonstrated by targeted disruption of the HPRT1 gene to create isogenic disease models of Lesch-Nyhan-Syndrome. Using female hPSCs, homozygous disruption of HPRT1 occurred at efficiencies of up to 15%. Differentiating isogenic knock-out cells both into central nervous system (CNS) as well as into sensory-like neurons recapitulated previously described phenotypes based on patient-specific induced PSCs and extended these findings to non-CNS neurons, respectively. CONCLUSION: The combined vector system allows for flexible and affordable generation of knock-out hPSCs lines, thus enabling investigation of developmental processes as well as the generation of isogenic disease models without the need for patient material.


Asunto(s)
Desoxirribonucleasas de Localización Especificada Tipo II/genética , Endonucleasas/genética , Hipoxantina Fosforribosiltransferasa/biosíntesis , Síndrome de Lesch-Nyhan/genética , Células Madre Pluripotentes/metabolismo , Proteínas Recombinantes de Fusión/genética , Transactivadores/genética , Endonucleasas/metabolismo , Femenino , Técnicas de Inactivación de Genes , Marcación de Gen , Genómica , Homocigoto , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Síndrome de Lesch-Nyhan/metabolismo , Síndrome de Lesch-Nyhan/patología , Células Madre Pluripotentes/citología , Células Receptoras Sensoriales/metabolismo
19.
Mol Genet Metab ; 110(3): 268-74, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24075303

RESUMEN

We describe a family of seven boys affected by Lesch-Nyhan disease with various phenotypes. Further investigations revealed a mutation c.203T>C in the gene encoding HGprt of all members, with substitution of leucine to proline at residue 68 (p.Leu68Pro). Thus patients from this family display a wide variety of symptoms although sharing the same mutation. Mutant HGprt enzyme was prepared by site-directed mutagenesis and the kinetics of the enzyme revealed that the catalytic activity of the mutant was reduced, in association with marked reductions in the affinity towards phosphoribosylpyrophosphate (PRPP). Its Km for PRPP was increased 215-fold with hypoxanthine as substrate and 40-fold with guanine as substrate with associated reduced catalytic potential. Molecular modeling confirmed that the most prominent defect was the dramatically reduced affinity towards PRPP. Our studies suggest that the p.Leu68Pro mutation has a strong impact on PRPP binding and on stability of the active conformation. This suggests that factors other than HGprt activity per se may influence the phenotype of Lesch-Nyhan patients.


Asunto(s)
Hipoxantina Fosforribosiltransferasa/deficiencia , Hipoxantina Fosforribosiltransferasa/genética , Fenotipo , Adolescente , Adulto , Sustitución de Aminoácidos , Niño , Codón , Activación Enzimática , Humanos , Hipoxantina Fosforribosiltransferasa/química , Cinética , Síndrome de Lesch-Nyhan/diagnóstico , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/metabolismo , Masculino , Persona de Mediana Edad , Modelos Moleculares , Mutación , Linaje , Unión Proteica , Conformación Proteica , Estabilidad Proteica , Adulto Joven
20.
J Neural Transm (Vienna) ; 120(9): 1359-67, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23400363

RESUMEN

In the present study, we investigate the in vitro effect of hypoxanthine on acetylcholinesterase and butyrylcholinesterase activities in the hippocampus, striatum, cerebral cortex and serum of 15-, 30- and 60-day-old rats. Furthermore, we also evaluated the influence of antioxidants, namely α-tocopherol (trolox) and ascorbic acid, and allopurinol to investigate the possible participation of free radicals and uric acid in the effects elicited by hypoxanthine on these parameters. Acetylcholinesterase and butyrylcholinesterase activities were determined according to Ellman et al. (Biochem Pharmacol 7:88-95, 1961), with some modifications. Hypoxanthine (10.0 µM), when added to the incubation medium, enhanced acetylcholinesterase activity in the hippocampus and striatum of 15- and 30-day-old rats and reduced butyrylcholinesterase activity in the serum of 60-day-old rats. The administration of allopurinol and/or antioxidants partially prevented the alterations caused by hypoxanthine in acetylcholinesterase and butyrylcholinesterase activities in the cerebrum and serum of rats. Data indicate that hypoxanthine alters cholinesterase activities, probably through free radicals and uric acid production since the alterations were prevented by the administration of allopurinol and antioxidants. It is presumed that the cholinesterase system may be associated, at least in part, with the neuronal dysfunction observed in patients affected by Lesch-Nyhan disease. In addition, although extrapolation of findings from animal experiments to humans is difficult, it is conceivable that these vitamins and allopurinol might serve as an adjuvant therapy to avoid progression of brain damage in patients affected by this disease.


Asunto(s)
Alopurinol/farmacología , Antioxidantes/farmacología , Colinesterasas/metabolismo , Inhibidores Enzimáticos/farmacología , Hipoxantina/farmacología , Acetilcolinesterasa/metabolismo , Análisis de Varianza , Animales , Ácido Ascórbico/farmacología , Butirilcolinesterasa/metabolismo , Radicales Libres/metabolismo , Hipoxantina/líquido cefalorraquídeo , Síndrome de Lesch-Nyhan/metabolismo , Ratas , Ratas Wistar , Ácido Úrico/metabolismo , alfa-Tocoferol/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA