Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 230
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 17(2): e1008859, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33539341

RESUMEN

Abnormal protein aggregation within neurons is a key pathologic feature of Parkinson's disease (PD). The spread of brain protein aggregates is associated with clinical disease progression, but how this occurs remains unclear. Mutations in glucosidase, beta acid 1 (GBA), which encodes glucocerebrosidase (GCase), are the most penetrant common genetic risk factor for PD and dementia with Lewy bodies and associate with faster disease progression. To explore how GBA mutations influence pathogenesis, we previously created a Drosophila model of GBA deficiency (Gba1b) that manifests neurodegeneration and accelerated protein aggregation. Proteomic analysis of Gba1b mutants revealed dysregulation of proteins involved in extracellular vesicle (EV) biology, and we found altered protein composition of EVs from Gba1b mutants. Accordingly, we hypothesized that GBA may influence pathogenic protein aggregate spread via EVs. We found that accumulation of ubiquitinated proteins and Ref(2)P, Drosophila homologue of mammalian p62, were reduced in muscle and brain tissue of Gba1b flies by ectopic expression of wildtype GCase in muscle. Neuronal GCase expression also rescued protein aggregation both cell-autonomously in brain and non-cell-autonomously in muscle. Muscle-specific GBA expression reduced the elevated levels of EV-intrinsic proteins and Ref(2)P found in EVs from Gba1b flies. Perturbing EV biogenesis through neutral sphingomyelinase (nSMase), an enzyme important for EV release and ceramide metabolism, enhanced protein aggregation when knocked down in muscle, but did not modify Gba1b mutant protein aggregation when knocked down in neurons. Lipidomic analysis of nSMase knockdown on ceramide and glucosylceramide levels suggested that Gba1b mutant protein aggregation may depend on relative depletion of specific ceramide species often enriched in EVs. Finally, we identified ectopically expressed GCase within isolated EVs. Together, our findings suggest that GCase deficiency promotes accelerated protein aggregate spread between cells and tissues via dysregulated EVs, and EV-mediated trafficking of GCase may partially account for the reduction in aggregate spread.


Asunto(s)
Drosophila melanogaster/metabolismo , Vesículas Extracelulares/metabolismo , Glucosilceramidasa/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Agregación Patológica de Proteínas/metabolismo , Animales , Transporte Biológico , Encéfalo/metabolismo , Ceramidas/metabolismo , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Técnicas de Silenciamiento del Gen , Glucosilceramidasa/deficiencia , Glucosilceramidasa/genética , Glucosilceramidas/metabolismo , Lipidómica , Músculos/metabolismo , Mutación , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Agregación Patológica de Proteínas/genética , Proteoma/genética , Proteoma/metabolismo , Interferencia de ARN
2.
Int J Mol Sci ; 25(12)2024 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-38928321

RESUMEN

Glucocerebrosidase (GCase) is a lysosomal enzyme that catalyzes the breakdown of glucosylceramide in the presence of its activator saposin C (SapC). SapC arises from the proteolytical cleavage of prosaposin (encoded by PSAP gene), which gives rise to four saposins. GCase is targeted to the lysosomes by LIMP-2, encoded by SCARB2 gene. GCase deficiency causes Gaucher Disease (GD), which is mainly due to biallelic pathogenetic variants in the GCase-encoding gene, GBA1. However, impairment of GCase activity can be rarely caused by SapC or LIMP-2 deficiencies. We report a new case of LIMP-2 deficiency and a new case of SapC deficiency (missing all four saposins, PSAP deficiency), and measured common biomarkers of GD and GCase activity. Glucosylsphingosine and chitotriosidase activity in plasma were increased in GCase deficiencies caused by PSAP and GBA1 mutations, whereas SCARB2-linked deficiency showed only Glucosylsphingosine elevation. GCase activity was reduced in fibroblasts and leukocytes: the decrease was sharper in GBA1- and SCARB2-mutant fibroblasts than PSAP-mutant ones; LIMP-2-deficient leukocytes displayed higher residual GCase activity than GBA1-mutant ones. Finally, we demonstrated that GCase mainly undergoes proteasomal degradation in LIMP-2-deficient fibroblasts and lysosomal degradation in PSAP-deficient fibroblasts. Thus, we analyzed the differential biochemical profile of GCase deficiencies due to the ultra-rare PSAP and SCARB2 biallelic pathogenic variants in comparison with the profile observed in GBA1-linked GCase deficiency.


Asunto(s)
Enfermedad de Gaucher , Glucosilceramidasa , Proteínas de Membrana de los Lisosomas , Receptores Depuradores , Saposinas , Glucosilceramidasa/genética , Glucosilceramidasa/deficiencia , Glucosilceramidasa/metabolismo , Humanos , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/metabolismo , Saposinas/deficiencia , Saposinas/genética , Saposinas/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Proteínas de Membrana de los Lisosomas/genética , Receptores Depuradores/genética , Receptores Depuradores/metabolismo , Fibroblastos/metabolismo , Mutación , Lisosomas/metabolismo , Lisosomas/enzimología , Hexosaminidasas/metabolismo , Hexosaminidasas/genética , Hexosaminidasas/deficiencia , Masculino , Femenino
3.
Nature ; 543(7643): 108-112, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28225753

RESUMEN

Gaucher disease is caused by mutations in GBA1, which encodes the lysosomal enzyme glucocerebrosidase (GCase). GBA1 mutations drive extensive accumulation of glucosylceramide (GC) in multiple innate and adaptive immune cells in the spleen, liver, lung and bone marrow, often leading to chronic inflammation. The mechanisms that connect excess GC to tissue inflammation remain unknown. Here we show that activation of complement C5a and C5a receptor 1 (C5aR1) controls GC accumulation and the inflammatory response in experimental and clinical Gaucher disease. Marked local and systemic complement activation occurred in GCase-deficient mice or after pharmacological inhibition of GCase and was associated with GC storage, tissue inflammation and proinflammatory cytokine production. Whereas all GCase-inhibited mice died within 4-5 weeks, mice deficient in both GCase and C5aR1, and wild-type mice in which GCase and C5aR were pharmacologically inhibited, were protected from these adverse effects and consequently survived. In mice and humans, GCase deficiency was associated with strong formation of complement-activating GC-specific IgG autoantibodies, leading to complement activation and C5a generation. Subsequent C5aR1 activation controlled UDP-glucose ceramide glucosyltransferase production, thereby tipping the balance between GC formation and degradation. Thus, extensive GC storage induces complement-activating IgG autoantibodies that drive a pathway of C5a generation and C5aR1 activation that fuels a cycle of cellular GC accumulation, innate and adaptive immune cell recruitment and activation in Gaucher disease. As enzyme replacement and substrate reduction therapies are expensive and still associated with inflammation, increased risk of cancer and Parkinson disease, targeting C5aR1 may serve as a treatment option for patients with Gaucher disease and, possibly, other lysosomal storage diseases.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Enfermedad de Gaucher/inmunología , Enfermedad de Gaucher/patología , Glucosilceramidas/inmunología , Glucosilceramidas/metabolismo , Inflamación/inmunología , Inflamación/patología , Animales , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Autoanticuerpos/inmunología , Activación de Complemento , Complemento C5a/biosíntesis , Complemento C5a/inmunología , Proteínas del Sistema Complemento/biosíntesis , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Enfermedad de Gaucher/metabolismo , Enfermedad de Gaucher/prevención & control , Glucosilceramidasa/antagonistas & inhibidores , Glucosilceramidasa/deficiencia , Glucosilceramidasa/genética , Glucosiltransferasas/biosíntesis , Glucosiltransferasas/metabolismo , Humanos , Inmunoglobulina G/inmunología , Inflamación/metabolismo , Inflamación/prevención & control , Masculino , Ratones , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/inmunología , Receptor de Anafilatoxina C5a/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología
4.
Ann Neurol ; 90(3): 490-505, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34288055

RESUMEN

OBJECTIVE: We utilized human midbrain-like organoids (hMLOs) generated from human pluripotent stem cells carrying glucocerebrosidase gene (GBA1) and α-synuclein (α-syn; SNCA) perturbations to investigate genotype-to-phenotype relationships in Parkinson disease, with the particular aim of recapitulating α-syn- and Lewy body-related pathologies and the process of neurodegeneration in the hMLO model. METHODS: We generated and characterized hMLOs from GBA1-/- and SNCA overexpressing isogenic embryonic stem cells and also generated Lewy body-like inclusions in GBA1/SNCA dual perturbation hMLOs and conduritol-b-epoxide-treated SNCA triplication hMLOs. RESULTS: We identified for the first time that the loss of glucocerebrosidase, coupled with wild-type α-syn overexpression, results in a substantial accumulation of detergent-resistant, ß-sheet-rich α-syn aggregates and Lewy body-like inclusions in hMLOs. These Lewy body-like inclusions exhibit a spherically symmetric morphology with an eosinophilic core, containing α-syn with ubiquitin, and can also be formed in Parkinson disease patient-derived hMLOs. We also demonstrate that impaired glucocerebrosidase function promotes the formation of Lewy body-like inclusions in hMLOs derived from patients carrying the SNCA triplication. INTERPRETATION: Taken together, the data indicate that our hMLOs harboring 2 major risk factors (glucocerebrosidase deficiency and wild-type α-syn overproduction) of Parkinson disease provide a tractable model to further elucidate the underlying mechanisms for progressive Lewy body formation. ANN NEUROL 2021;90:490-505.


Asunto(s)
Glucosilceramidasa/deficiencia , Cuerpos de Lewy/metabolismo , Mesencéfalo/metabolismo , Mutación/fisiología , Organoides/metabolismo , alfa-Sinucleína/biosíntesis , Células Madre Embrionarias/metabolismo , Glucosilceramidasa/genética , Humanos , Cuerpos de Lewy/genética , Cuerpos de Lewy/patología , Mesencéfalo/patología , Organoides/patología , alfa-Sinucleína/genética
5.
Am J Med Genet A ; 185(8): 2471-2476, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34031990

RESUMEN

Gaucher disease (GD), one of the most common lysosomal disorders, is caused by deficiency of ß-glucocerebrosidase. Based on the presence and severity of neurological complications, GD is classified into types I, II (the most severe form), and III. Abnormalities in systemic markers of vitamin B12 (B12 ) metabolism have been reported in GD type I patients, suggesting a higher prevalence of B12 deficiency in these patients. A 2-month-old male with GD type II was admitted to the hospital presenting jaundice, hepatosplenomegaly, and ichthyosis. At admission, cholestasis and ascites, abnormal liver function enzymes, prolonged prothrombin time, and high levels of B12 were confirmed. Analysis of biomarkers of B12 status revealed elevated B12 and holo-transcobalamin (holo-TC) levels. The B12 profile found in our patient is the opposite to what is described for GD type I patients. Holo-TC may increase in inflammatory states or due to liver diseases. In GD, the accumulation of glucocerebroside may be a trigger that initiates a systemic inflammatory reaction, characterized by macrophage activation. We suggest higher levels of holo-TC could be associated with a more severe (neuronopathic) GD, and be a biomarker of GD type II.


Asunto(s)
Biomarcadores/sangre , Enfermedad de Gaucher/sangre , Enfermedad de Gaucher/diagnóstico , Transcobalaminas , Enfermedad de Gaucher/genética , Glucosilceramidasa/deficiencia , Glucosilceramidasa/genética , Humanos , Lactante , Masculino , Pronóstico , Evaluación de Síntomas , Transcobalaminas/metabolismo , Vitamina B 12/metabolismo
6.
Am J Hematol ; 96(5): 545-551, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33606887

RESUMEN

Ambroxol hydrochloride is an oral mucolytic drug available over-the-counter for many years as cough medicine. In 2009 it was identified as a pharmacological chaperone for mutant glucocerebrosidase, albeit in a several-fold higher dose. Unfortunately, there have been no pharma-driven clinical trials to establish its use. Thus, real-world observational data are needed on the safety and efficacy of ambroxol for patients with Gaucher disease (GD) and GBA-Parkinson disease (GBA-PD). Clinicians treating patients with ambroxol for GD and GBA-PD were approached to collaborate in an investigator-initiated registry. Anonymized data were collected, including demographics, GD type, GD-specific therapy (when applicable), adverse events (AEs), and, when available, efficacy data. We report the data of the first 41 patients (25 females) at a median (range) age 17 (1.5-74) from 13 centers; 11 with GD type 1(four diagnosed with PD), 27 with neuronopathic GD (nGD), and three GBA mutation carriers with PD. The median (range) treatment period and maximum dose of ambroxol were 19 (1-76) months and 435 (75-1485) mg/day, respectively. One patient with type 2 GD died of her disease. No other severe AEs were reported. Twelve patients experienced AE, including minor bowel discomfort, cough, allergic reaction, mild proteinuria, dizziness and disease progression. Clinical benefits were reported in 25 patients, including stable or improved neurological status, increased physical activity, and reduced fatigue. Until the approval of specific therapies for nGD and disease-modification for GBA-PD, these preliminary data may be encouraging to physicians and patients who consider an off-label use of ambroxol.


Asunto(s)
Ambroxol/uso terapéutico , Enfermedad de Gaucher/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico , Sistema de Registros , Adolescente , Adulto , Anciano , Ambroxol/efectos adversos , Ambroxol/farmacología , Disponibilidad Biológica , Barrera Hematoencefálica , Niño , Preescolar , Terapia Combinada , Terapia de Reemplazo Enzimático , Femenino , Glucosilceramidasa/deficiencia , Glucosilceramidasa/genética , Glucosilceramidasa/metabolismo , Glucosilceramidasa/uso terapéutico , Humanos , Lactante , Masculino , Persona de Mediana Edad , Uso Fuera de lo Indicado , Enfermedad de Parkinson/genética , Estabilidad Proteica/efectos de los fármacos , Adulto Joven
7.
Proc Natl Acad Sci U S A ; 115(4): 798-803, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311330

RESUMEN

Accumulating evidence suggests that α-synuclein (α-syn) occurs physiologically as a helically folded tetramer that resists aggregation. However, the mechanisms underlying the regulation of formation of α-syn tetramers are still mostly unknown. Cellular membrane lipids are thought to play an important role in the regulation of α-syn tetramer formation. Since glucocerebrosidase 1 (GBA1) deficiency contributes to the aggregation of α-syn and leads to changes in neuronal glycosphingolipids (GSLs) including gangliosides, we hypothesized that GBA1 deficiency may affect the formation of α-syn tetramers. Here, we show that accumulation of GSLs due to GBA1 deficiency decreases α-syn tetramers and related multimers and increases α-syn monomers in CRISPR-GBA1 knockout (KO) SH-SY5Y cells. Moreover, α-syn tetramers and related multimers are decreased in N370S GBA1 Parkinson's disease (PD) induced pluripotent stem cell (iPSC)-derived human dopaminergic (hDA) neurons and murine neurons carrying the heterozygous L444P GBA1 mutation. Treatment with miglustat to reduce GSL accumulation and overexpression of GBA1 to augment GBA1 activity reverse the destabilization of α-syn tetramers and protect against α-syn preformed fibril-induced toxicity in hDA neurons. Taken together, these studies provide mechanistic insights into how GBA1 regulates the transition from monomeric α-syn to α-syn tetramers and multimers and suggest unique therapeutic opportunities for PD and dementia with Lewy bodies.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Glucosilceramidasa/deficiencia , Glicoesfingolípidos/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , 1-Desoxinojirimicina/análogos & derivados , Línea Celular Tumoral , Glucosilceramidasa/genética , Humanos , Multimerización de Proteína
8.
PLoS Genet ; 14(9): e1007694, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30256786

RESUMEN

Mutations in the glucosylceramidase beta (GBA) gene are strongly associated with neurodegenerative diseases marked by protein aggregation. GBA encodes the lysosomal enzyme glucocerebrosidase, which breaks down glucosylceramide. A common explanation for the link between GBA mutations and protein aggregation is that lysosomal accumulation of glucosylceramide causes impaired autophagy. We tested this hypothesis directly by measuring protein turnover and abundance in Drosophila mutants with deletions in the GBA ortholog Gba1b. Proteomic analyses revealed that known autophagy substrates, which had severely impaired turnover in autophagy-deficient Atg7 mutants, showed little to no overall slowing of turnover or increase in abundance in Gba1b mutants. Likewise, Gba1b mutants did not have the marked impairment of mitochondrial protein turnover seen in mitophagy-deficient parkin mutants. Proteasome activity, microautophagy, and endocytic degradation also appeared unaffected in Gba1b mutants. However, we found striking changes in the turnover and abundance of proteins associated with extracellular vesicles (EVs), which have been proposed as vehicles for the spread of protein aggregates in neurodegenerative disease. These changes were specific to Gba1b mutants and did not represent an acceleration of normal aging. Western blotting of isolated EVs confirmed the increased abundance of EV proteins in Gba1b mutants, and nanoparticle tracking analysis revealed that Gba1b mutants had six times as many EVs as controls. Genetic perturbations of EV production in Gba1b mutants suppressed protein aggregation, demonstrating that the increase in EV abundance contributed to the accumulation of protein aggregates. Together, our findings indicate that glucocerebrosidase deficiency causes pathogenic changes in EV metabolism and may promote the spread of protein aggregates through extracellular vesicles.


Asunto(s)
Proteínas de Drosophila/genética , Vesículas Extracelulares/patología , Glucosilceramidasa/deficiencia , Enfermedad de Parkinson/patología , Agregación Patológica de Proteínas/patología , Animales , Animales Modificados Genéticamente , Autofagia/genética , Proteína 7 Relacionada con la Autofagia/genética , Modelos Animales de Enfermedad , Drosophila , Femenino , Glucosilceramidasa/genética , Humanos , Masculino , Mutación , Enfermedad de Parkinson/genética , Agregación Patológica de Proteínas/genética , Proteómica
9.
Int J Mol Sci ; 22(23)2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34884544

RESUMEN

Parkinson's Disease (PD) is the most common movement disorder, and the strongest genetic risk factor for PD is mutations in the glucocerebrosidase gene (GBA). Mutations in GBA also lead to the development of Gaucher Disease (GD), the most common type of lysosomal storage disorder. Current therapeutic approaches fail to address neurological GD symptoms. Therefore, identifying therapeutic strategies that improve the phenotypic traits associated with GD/PD in animal models may provide an opportunity for treating neurological manifestations of GD/PD. Thiazolidinediones (TZDs, also called glitazones) are a class of compounds targeted for the treatment of type 2 diabetes, and have also shown promise for the treatment of neurodegenerative disease, including PD. Here, we tested the efficacy of glitazone administration during development in a fly GD model with deletions in the GBA homolog, dGBA1b (GBA1ΔTT/ΔTT). We observed an optimal dose of pioglitazone (PGZ) at a concentration of 1 µM that reduced sleep deficits, locomotor impairments, climbing defects, and restoration of normal protein levels of Ref(2)P, a marker of autophagic flux, in GBA1ΔTT/ΔTT mutant flies, compared to GBA1+/+ control flies. These data suggest that PGZ may represent a potential compound with which to treat GD/PD by improving function of lysosomal-autophagy pathways, a cellular process that removes misfolded or aggregated proteins.


Asunto(s)
Enfermedad de Gaucher/tratamiento farmacológico , Glucosilceramidasa/deficiencia , Enfermedad de Parkinson/tratamiento farmacológico , Tiazolidinedionas/farmacología , Animales , Drosophila melanogaster , Enfermedad de Gaucher/etiología , Enfermedad de Gaucher/patología , Humanos , Masculino , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Fenotipo
10.
Mol Genet Metab ; 130(4): 274-282, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32536424

RESUMEN

Gaucher disease (GD) is caused by pathogenic mutations in GBA1, the gene that encodes the lysosomal enzyme ß-glucocerebrosidase. Until now, treatments for GD cannot completely reverse bone problems. The aim of this work was to evaluate the potential of MSCs from GD patients (GD MSCs) to differentiate towards the osteoblast (GD Ob) and adipocyte (GD Ad) lineages, and their role in osteoclastogenesis. We observed that GD Ob exhibited reduced mineralization, collagen deposition and alkaline phosphatase activity (ALP), as well as decreased gene expression of RUNX2, COLA1 and ALP. We also evaluated the process of osteoclastogenesis and observed that conditioned media from GD MSCs supernatants induced an increase in the number of osteoclasts. In this model, osteoclastogenesis was induced by RANKL and IL-1ß. Furthermore, results showed that in GD MSCs there was a promotion in NLRP3 and PPAR-γ gene expression. Adipogenic differentiation revealed that GD Ad had an increase in PPAR-γ and a reduced RUNX2 gene expression, promoting adipocyte differentiation. In conclusion, our results show that GD MSCs exhibited deficient GD Ob differentiation and increased adipogenesis. In addition, we show that GD MSCs promoted increased osteoclastogenesis through RANKL and IL-1ß. These changes in GD MSCs are likely to contribute to skeletal imbalance observed in GD patients.


Asunto(s)
Adipogénesis , Diferenciación Celular , Enfermedad de Gaucher/patología , Glucosilceramidasa/deficiencia , Células Madre Mesenquimatosas/patología , Osteoclastos/patología , Osteogénesis , Apoptosis , Ciclo Celular , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Enfermedad de Gaucher/metabolismo , Regulación de la Expresión Génica , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoclastos/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo
11.
J Inherit Metab Dis ; 43(5): 1056-1059, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32242941

RESUMEN

Neuronopathic Gaucher disease (nGD) has a very wide clinical and genotypic spectrum. However, there is no consensus definition of nGD, including no description of how best to diagnostically separate the acute form-Gaucher type 2-from the subacute or chronic form-Gaucher type 3. In this article, we define the various forms of Gaucher disease with particular emphasis on the presence of gaze palsy in all patients with nGD. This consensus definition will help in both clinical diagnosis and appropriate patient recruitment to upcoming clinical trials.


Asunto(s)
Enfermedad de Gaucher/diagnóstico , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/fisiopatología , Genotipo , Glucosilceramidasa/deficiencia , Humanos , Oftalmoplejía/etiología , Terminología como Asunto
12.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-32998334

RESUMEN

The challenges in the diagnosis, prognosis, and monitoring of Gaucher disease (GD), an autosomal recessive inborn error of glycosphingolipid metabolism, can negatively impact clinical outcomes. This systematic literature review evaluated the value of glucosylsphingosine (lyso-Gb1), as the most reliable biomarker currently available for the diagnosis, prognosis, and disease/treatment monitoring of patients with GD. Literature searches were conducted using MEDLINE, Embase, PubMed, ScienceOpen, Science.gov, Biological Abstracts, and Sci-Hub to identify original research articles relevant to lyso-Gb1 and GD published before March 2019. Seventy-four articles met the inclusion criteria, encompassing 56 related to pathology and 21 related to clinical biomarkers. Evidence for lyso-Gb1 as a pathogenic mediator of GD was unequivocal, although its precise role requires further elucidation. Lyso-Gb1 was deemed a statistically reliable diagnostic and pharmacodynamic biomarker in GD. Evidence supports lyso-Gb1 as a disease-monitoring biomarker for GD, and some evidence supports lyso-Gb1 as a prognostic biomarker, but further study is required. Lyso-Gb1 meets the criteria for a biomarker as it is easily accessible and reliably quantifiable in plasma and dried blood spots, enables the elucidation of GD molecular pathogenesis, is diagnostically valuable, and reflects therapeutic responses. Evidentiary standards appropriate for verifying inter-laboratory lyso-Gb1 concentrations in plasma and in other anatomical sites are needed.


Asunto(s)
Enfermedad de Gaucher/sangre , Glucosilceramidasa/deficiencia , Lisosomas/metabolismo , Psicosina/análogos & derivados , Biomarcadores/sangre , Encéfalo/metabolismo , Encéfalo/patología , Cromatografía Líquida de Alta Presión , Enfermedad de Gaucher/diagnóstico , Enfermedad de Gaucher/tratamiento farmacológico , Enfermedad de Gaucher/patología , Expresión Génica , Glucosilceramidasa/genética , Glucosilceramidasa/uso terapéutico , Humanos , Hígado/metabolismo , Hígado/patología , Lisosomas/enzimología , Lisosomas/patología , Monitoreo Fisiológico/métodos , Psicosina/sangre , Bazo/metabolismo , Bazo/patología , Espectrometría de Masas en Tándem
13.
J Lipid Res ; 60(11): 1851-1867, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31562193

RESUMEN

ß-glucosidases [GBA1 (glucocerebrosidase) and GBA2] are ubiquitous essential enzymes. Lysosomal GBA1 and cytosol-facing GBA2 degrade glucosylceramide (GlcCer); GBA1 deficiency causes Gaucher disease, a lysosomal storage disorder characterized by lysosomal accumulation of GlcCer, which is partly converted to glucosylsphingosine (GlcSph). GBA1 and GBA2 also may transfer glucose from GlcCer to cholesterol, yielding glucosylated cholesterol (GlcChol). Here, we aimed to clarify the role of zebrafish Gba2 in glycosphingolipid metabolism during Gba1 deficiency in zebrafish (Danio rerio), which are able to survive total Gba1 deficiency. We developed Gba1 (gba1-/-), Gba2 (gba2-/-), and double (gba1-/-:gba2-/-) zebrafish knockouts using CRISPR/Cas9 and explored the effects of both genetic and pharmacological interventions on GlcCer metabolism in individual larvae. Activity-based probes and quantification of relevant glycolipid metabolites confirmed enzyme deficiency. GlcSph increased in gba1-/- larvae (0.09 pmol/fish) but did not increase more in gba1-/-:gba2-/- larvae. GlcCer was comparable in gba1-/- and WT larvae but increased in gba2-/- and gba1-/-:gba2-/- larvae. Independent of Gba1 status, GlcChol was low in all gba2-/- larvae (0.05 vs. 0.18 pmol/fish in WT). Pharmacologic inactivation of zebrafish Gba1 comparably increased GlcSph. Inhibition of GlcCer synthase (GCS) in Gba1-deficient larvae reduced GlcCer and GlcSph, and concomitant inhibition of GCS and Gba2 with iminosugars also reduced excessive GlcChol. Finally, overexpression of human GBA1 and injection of recombinant GBA1 both decreased GlcSph. We determined that zebrafish larvae offer an attractive model to study glucosidase actions in glycosphingolipid metabolism in vivo, and we identified distinguishing characteristics of zebrafish Gba2 deficiency.


Asunto(s)
Glucosilceramidasa/deficiencia , Glicoesfingolípidos/metabolismo , Modelos Biológicos , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo , beta-Glucosidasa/metabolismo , Animales , Células Cultivadas , Glucosilceramidasa/metabolismo , Pez Cebra , beta-Glucosidasa/deficiencia
14.
Am J Hematol ; 94(1): 29-38, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30264864

RESUMEN

Eliglustat is a first-line oral therapy for adults with Gaucher disease type 1 (GD1) and poor, intermediate or extensive CYP2D6-metabolizer phenotypes (>90% of patients). We report the final results of a Phase 2 trial and extension (NCT00358150) in previously untreated adult GD1 patients who had splenomegaly with thrombocytopenia and/or anemia and received 50 or 100 mg eliglustat tartrate (equivalent to 42 or 84 mg eliglustat) twice daily for 8 years. In total, 19 of 26 patients completed the trial. After 8 years of eliglustat, mean spleen and liver volumes decreased by 69% and 34%, respectively. Mean hemoglobin concentration and platelet count increased by 2.2 g/dL and 113%, respectively. All patients met at least 3 of 4 therapeutic goals established for patients on long-term enzyme replacement therapy. Mean final values for patients with severe splenomegaly (n = 6), moderate-to-severe anemia (n = 6), or severe thrombocytopenia (n = 8) were similar to patients with milder disease at baseline and within long-term therapeutic goal thresholds. Biomarker median percent changes from baseline were -91% for chitotriosidase, -87% for CCL18, -92% for glucosylsphingosine, and -80% for plasma glucosylceramide. Mean lumbar spine T-score increased by 0.96, moving from the osteopenic to the normal range. Mean quality-of-life scores, mostly below normal at baseline, moved into ranges seen in healthy adults. Eliglustat was well-tolerated; 98% of adverse events were mild or moderate and 94% were considered unrelated to treatment. Clinically meaningful improvements in all parameters continued or were maintained over 8 years, with the largest margins of improvement seen in the most severely affected patients.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Enfermedad de Gaucher/tratamiento farmacológico , Glucosiltransferasas/antagonistas & inhibidores , Pirrolidinas/uso terapéutico , Adulto , Densidad Ósea , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Enfermedades Óseas Metabólicas/etiología , Femenino , Estudios de Seguimiento , Enfermedad de Gaucher/sangre , Enfermedad de Gaucher/complicaciones , Glucosilceramidasa/deficiencia , Enfermedades Hematológicas/sangre , Enfermedades Hematológicas/tratamiento farmacológico , Enfermedades Hematológicas/etiología , Hemoglobinas/análisis , Hepatomegalia/tratamiento farmacológico , Hepatomegalia/etiología , Hepatomegalia/patología , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Recuento de Plaquetas , Bazo/efectos de los fármacos , Bazo/patología , Esplenomegalia/tratamiento farmacológico , Esplenomegalia/etiología , Esplenomegalia/patología , Resultado del Tratamiento
15.
Brain ; 140(10): 2706-2721, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28969384

RESUMEN

Mutations in glucocerebrosidase 1 (GBA1) represent the most prevalent risk factor for Parkinson's disease. The molecular mechanisms underlying the link between GBA1 mutations and Parkinson's disease are incompletely understood. We analysed two aged (24-month-old) Gba1 mouse models, one carrying a knock-out mutation and the other a L444P knock-in mutation. A significant reduction of glucocerebrosidase activity was associated with increased total alpha-synuclein accumulation in both these models. Gba1 mutations alone did not alter the number of nigral dopaminergic neurons nor striatal dopamine levels. We then investigated the effect of overexpression of human alpha-synuclein in the substantia nigra of aged (18 to 21-month-old) L444P Gba1 mice. Following intraparenchymal injections of human alpha-synuclein carrying viral vectors, pathological accumulation of phosphorylated alpha-synuclein occurred within the transduced neurons. Stereological counts of nigral dopaminergic neurons revealed a significantly greater cell loss in Gba1-mutant than wild-type mice. These results indicate that Gba1 deficiency enhances neuronal vulnerability to neurodegenerative processes triggered by increased alpha-synuclein expression.


Asunto(s)
Dopamina/metabolismo , Glucosilceramidasa/genética , Mutación/genética , Neuronas/patología , Sustancia Negra/patología , alfa-Sinucleína/metabolismo , Factores de Edad , Animales , Encéfalo/metabolismo , Encéfalo/patología , Glucosilceramidasa/deficiencia , Humanos , Leucina/genética , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Prolina/genética , Desempeño Psicomotor/fisiología , Olfato/genética , Sustancia Negra/metabolismo , Transducción Genética , Tirosina 3-Monooxigenasa/metabolismo , beta-N-Acetilhexosaminidasas/metabolismo
16.
PLoS Genet ; 11(4): e1005065, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25835295

RESUMEN

Homozygous mutations in the glucocerebrosidase (GBA) gene result in Gaucher disease (GD), the most common lysosomal storage disease. Recent genetic studies have revealed that GBA mutations confer a strong risk for sporadic Parkinson's disease (PD). To investigate how GBA mutations cause PD, we generated GBA nonsense mutant (GBA-/-) medaka that are completely deficient in glucocerebrosidase (GCase) activity. In contrast to the perinatal death in humans and mice lacking GCase activity, GBA-/- medaka survived for months, enabling analysis of the pathological progression. GBA-/- medaka displayed the pathological phenotypes resembling human neuronopathic GD including infiltration of Gaucher cell-like cells into the brains, progressive neuronal loss, and microgliosis. Detailed pathological findings represented lysosomal abnormalities in neurons and alpha-synuclein (α-syn) accumulation in axonal swellings containing autophagosomes. Unexpectedly, disruption of α-syn did not improve the life span, formation of axonal swellings, neuronal loss, or neuroinflammation in GBA-/- medaka. Taken together, the present study revealed GBA-/- medaka as a novel neuronopathic GD model, the pahological mechanisms of α-syn accumulation caused by GCase deficiency, and the minimal contribution of α-syn to the pathogenesis of neuronopathic GD.


Asunto(s)
Axones/metabolismo , Enfermedad de Gaucher/genética , Glucosilceramidasa/deficiencia , Oryzias/genética , alfa-Sinucleína/metabolismo , Animales , Axones/ultraestructura , Modelos Animales de Enfermedad , Enfermedad de Gaucher/metabolismo , Enfermedad de Gaucher/patología , Glucosilceramidasa/genética , Oryzias/metabolismo , Fagosomas/metabolismo
17.
J Natl Med Assoc ; 110(4): 330-333, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30126557

RESUMEN

Gaucher disease is a lipid storage disorder due to deficiency of beta glucocerebrosidase. It's an autosomal recessive disease and as a result of this enzyme deficiency, glucocerebroside accumulates in various types of tissues like liver, brain spleen and bone marrow. We aimed to describe the effects of enzyme replacement therapy in three members of a family with Gaucher disease and to emphasize screening of the family members of the patients with Gaucher disease. Furthermore, late diagnosis and treatment in these patients have a minimal effect on improvement of the quality of life, and early diagnosis and treatment are very important in Gaucher disease.


Asunto(s)
Terapia de Reemplazo Enzimático , Enfermedad de Gaucher/tratamiento farmacológico , Glucosilceramidasa/uso terapéutico , Adolescente , Análisis Mutacional de ADN , Enfermedad de Gaucher/complicaciones , Enfermedad de Gaucher/genética , Glucosilceramidasa/deficiencia , Hemoglobinas/análisis , Humanos , Masculino , Persona de Mediana Edad , Esplenomegalia/etiología
18.
Georgian Med News ; (Issue): 69-73, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29578427

RESUMEN

The data on occurrence and pathogenetic mechanisms of glucosylceramide lipidosis (Gaucher disease), which is based on the hereditary deficiency of glucocerebrosidase activity -the enzyme involved in the processing of cellular metabolism products, is presented. Clinical and morphological manifestations of the disease, therapeutic tactics and prognosis are defined. A typical clinical case of Gaucher disease type 1 in two adult patients was analyzed. It is shown that timely diagnosis of Gaucher disease allows the prescription of adequate treatment by means of enzyme replacement therapy with cerezyme drug. That is why the correct and timely diagnosis of the disease can prolongate patient's life for many years. In this case, the patient should be under the care of doctors (pediatricians, general physicians, hematologists). Continued treatment of Gaucher disease with cerezyme stabilizes the pathological process completely, decreases significant changes in bones and parenchymal organs, improving patients' lives considerably. For that reason, the sooner the adequate therapy is started, the more effective the results will be.


Asunto(s)
Terapia de Reemplazo Enzimático/métodos , Enfermedad de Gaucher/tratamiento farmacológico , Glucosilceramidasa/uso terapéutico , Adulto , Enfermedad de Gaucher/enzimología , Enfermedad de Gaucher/fisiopatología , Glucosilceramidasa/deficiencia , Humanos , Masculino , Resultado del Tratamiento
19.
J Cell Physiol ; 232(12): 3454-3467, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28098348

RESUMEN

Lysosomal storage disorders (LDS) comprise a group of rare multisystemic diseases resulting from inherited gene mutations that impair lysosomal homeostasis. The most common LSDs, Gaucher disease (GD), and Fabry disease (FD) are caused by deficiencies in the lysosomal glucocerebrosidase (GBA) and alpha-galactosidase A (GLA) enzymes, respectively. Given the systemic nature of enzyme deficiency, we hypothesized that the stem cell compartment of GD and FD patients might be also affected. Among stem cells, mesenchymal stem cells (MSCs) are a commonly investigated population given their role in hematopoiesis and the homeostatic maintenance of many organs and tissues. Since the impairment of MSC functions could pose profound consequences on body physiology, we evaluated whether GBA and GLA silencing could affect the biology of MSCs isolated from bone marrow and amniotic fluid. Those cell populations were chosen given the former's key role in organ physiology and the latter's intriguing potential as an alternative stem cell model for human genetic disease. Our results revealed that GBA and GLA deficiencies prompted cell cycle arrest along with the impairment of autophagic flux and an increase of apoptotic and senescent cell percentages. Moreover, an increase in ataxia-telangiectasia-mutated staining 1 hr after oxidative stress induction and a return to basal level at 48 hr, along with persistent gamma-H2AX staining, indicated that MSCs properly activated DNA repair signaling, though some damages remained unrepaired. Our data therefore suggest that MSCs with reduced GBA or GLA activity are prone to apoptosis and senescence due to impaired autophagy and DNA repair capacity.


Asunto(s)
Líquido Amniótico/citología , Células de la Médula Ósea/enzimología , Enfermedad de Fabry/enzimología , Enfermedad de Gaucher/enzimología , Glucosilceramidasa/deficiencia , Células Madre Mesenquimatosas/enzimología , Interferencia de ARN , alfa-Galactosidasa/metabolismo , Apoptosis , Autofagia , Células de la Médula Ósea/patología , Separación Celular , Células Cultivadas , Senescencia Celular , Niño , Reparación del ADN , Enfermedad de Fabry/genética , Enfermedad de Fabry/patología , Femenino , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/patología , Glucosilceramidasa/genética , Humanos , Células Madre Mesenquimatosas/patología , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Puntos de Control de la Fase S del Ciclo Celular , Transducción de Señal , Nicho de Células Madre , Transfección , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , alfa-Galactosidasa/genética
20.
Hum Mol Genet ; 24(23): 6675-86, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26362253

RESUMEN

Alpha-synuclein (αSyn) plays a central role in the pathogenesis of Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Recent multicenter genetic studies have revealed that mutations in the glucocerebrosidase 1 (GBA1) gene, which are responsible for Gaucher's disease, are strong risk factors for PD and DLB. However, the mechanistic link between the functional loss of glucocerebrosidase (GCase) and the toxicity of αSyn in vivo is not fully understood. In this study, we employed Drosophila models to examine the effect of GCase deficiency on the neurotoxicity of αSyn and its molecular mechanism. Behavioral and histological analyses showed that knockdown of the Drosophila homolog of GBA1 (dGBA1) exacerbates the locomotor dysfunction, loss of dopaminergic neurons and retinal degeneration of αSyn-expressing flies. This phenotypic aggravation was associated with the accumulation of proteinase K (PK)-resistant αSyn, rather than with changes in the total amount of αSyn, raising the possibility that glucosylceramide (GlcCer), a substrate of GCase, accelerates the misfolding of αSyn. Indeed, in vitro experiments revealed that GlcCer directly promotes the conversion of recombinant αSyn into the PK-resistant form, representing a toxic conformational change. Similar to dGBA1 knockdown, knockdown of the Drosophila homolog of ß-galactosidase (ß-Gal) also aggravated locomotor dysfunction of the αSyn flies, and its substrate GM1 ganglioside accelerated the formation of PK-resistant αSyn. Our findings suggest that the functional loss of GCase or ß-Gal promotes the toxic conversion of αSyn via aberrant interactions between αSyn and their substrate glycolipids, leading to the aggravation of αSyn-mediated neurodegeneration.


Asunto(s)
Glucosilceramidasa/genética , Trastornos Parkinsonianos/etiología , alfa-Sinucleína/metabolismo , Animales , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Endopeptidasa K/metabolismo , Técnicas de Silenciamiento del Gen , Glucosilceramidasa/deficiencia , Glucosilceramidasa/metabolismo , Glucosilceramidas/metabolismo , Humanos , Masculino , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/fisiopatología , Agregación Patológica de Proteínas , Pliegue de Proteína , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA