Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Aging (Albany NY) ; 16(5): 4591-4608, 2024 02 29.
Article in English | MEDLINE | ID: mdl-38428407

ABSTRACT

BACKGROUND: Excessive lipids accumulation and hepatocytes death are prominent characteristics of non-alcoholic fatty liver disease (NAFLD). Nonetheless, the precise pathophysiological mechanisms are not fully elucidated. METHODS: HepG2 cells stimulated with palmitic acids and rats fed with high-fat diet were used as models for NAFLD. The impact of Glucosylceramidase Beta 3 (GBA3) on fatty acid oxidation (FAO) was assessed using Seahorse metabolic analyzer. Lipid content was measured both in vitro and in vivo. To evaluate NAFLD progression, histological analysis was performed along with measurements of inflammatory factors and liver enzyme levels. Western blot and immunohistochemistry were employed to examine the activity levels of necroptosis. Flow cytometry and reactive oxygen species (ROS) staining were utilized to assess levels of oxidative stress. RESULTS: GBA3 promoted FAO and enhanced the mitochondrial membrane potential without affecting glycolysis. These reduced the lipid accumulation. Rats supplemented with GBA3 exhibited lower levels of inflammatory factors and liver enzymes, resulting in a slower progression of NAFLD. GBA3 overexpression reduced ROS and the ratio of cell apoptosis. Phosphorylation level was reduced in the essential mediator, MLKL, implicated in necroptosis. Mechanistically, as a transcriptional coactivator, GBA3 promoted the expression of Carnitine Palmitoyltransferase 2 (CPT2), which resulted in enhanced FAO. CONCLUSIONS: Increased FAO resulting from GBA3 reduced oxidative stress and the production of ROS, thereby inhibiting necroptosis and delaying the progression of NAFLD. Our research offers novel insights into the potential therapeutic applications of GBA3 and FAO in the management and treatment of NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Rats , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Liver/metabolism , Reactive Oxygen Species/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Glucosylceramidase , Lipid Metabolism , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Lipids
2.
J Chem Neuroanat ; 130: 102271, 2023 07.
Article in English | MEDLINE | ID: mdl-37019342

ABSTRACT

BACKGROUND: Neurodegenerative illnesses like Parkinson's and Alzheimer's are largely caused by the accumulation of aggregated proteins. Heat shock proteins (HSPs), which are molecular chaperons, have been linked with the modulation of ß-glucocerebrosidase (GCase) function encoded by GBA1 and Synucleinopathies. Herein, the chaperonic properties of African walnut ethanolic extract (WNE) in manganese-induced Parkinsonian neuropathology in the hippocampus was examined. METHODOLOGY: 48 adult male rats weighing 185 g ± 10 g were randomly assigned into 6 (A - F) groups (n = 8) and treated orally as follows: A-PBS (1 ml daily for 28 days), B-WNE (200 mg/kg daily for 28 days), C- WNE (400 mg/kg daily for 28 days), D-Mn (100 mg/kg daily for 28 days), E-Mn plus WNE (100 mg/kg Mn + 200 mg/kg WNE daily concomitantly for 28 days), F-Mn plus WNE (100 mg/kg Mn + 400 mg/kg WNE daily concomitantly for 28 days). RESULTS: Rats treated with WNE showed increased levels of HSP70 and HSP90 in comparison with the Mn-intoxicated group. GCase activity also increased significantly in animals treated with WNE. Our results further revealed the therapeutic tendencies of WNE against Mn toxicity by modulating oligomeric α-synuclein levels, redox activity, and glucose bioenergetics. Furthermore, immunohistochemical evaluation revealed reduced expression of neurofibrillary tangles, and reactive astrogliosis following WNE treatment. CONCLUSION: The ethanolic extract of African Walnut induced the activation of HSPs and increased the expression of GBA1 gene in the hippocampus. Activated heat shock proteins suppressed neurodegenerative changes due to Manganese toxicity. WNE was also shown to modulate neuroinflammatory, bioenergetics and neural redox balance in Parkinson-like neuropathology. This study was limited to the use of crude walnut extract and the evaluation of non-motor cascades of Parkinson's disease.


Subject(s)
Juglans , Parkinson Disease , Male , Rats , Animals , Parkinson Disease/metabolism , Juglans/metabolism , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Heat-Shock Proteins/metabolism , Manganese , alpha-Synuclein/metabolism , Hippocampus/metabolism , Plant Extracts/pharmacology
3.
Environ Technol ; 44(9): 1334-1350, 2023 Apr.
Article in English | MEDLINE | ID: mdl-34719354

ABSTRACT

Microalgae biomass contributes to effluent bioremediation. It is a concentrated source of nutrients and organic carbon, making it a potential alternative as a soil biostimulant. In this context, this study aimed to evaluate the soil application of microalgae biomass produced from the meat processing industry effluent treatment. The biomass was applied dry and as a mixture to demonstrate its potential to increase plant production and soil metabolic functions, analyzed short-term. Doses of 0.25%, 0.5%, 1%, and 2% biomass were applied in soils from (i) Horizon A: taken at a depth between 0 and 10 cm and; (ii) Horizon B: taken at a depth between 20 and 40 cm. Corn growth (Zea Mays L.), basal soil respiration, microbial biomass carbon, total organic carbon, ß-glucosidase, acid phosphatase, arylsulfatase, and urease enzymatic activity were evaluated in each sample. It is concluded that applying 2% microalgae biomass led to higher basal soil respiration, microbial biomass carbon, and ß-glucosidase, acid phosphatase, arylsulfatase enzymatic activity in both soils. On the other hand, boron may have contributed to urease activity reduction in Soil A. Although 2% biomass led to higher soils characteristics, that dose did not promote higher plant growth. Hence, considering that plant growth must be in line with changes in soil characteristics, the result that provided the higher plant shoot dry matter mass was by applying 0.55% biomass in both soils. Therefore, the application of microalgae biomass produced from a meat processing industry effluent treatment promoted a biologically active soil and boosted plant growth.


Subject(s)
Microalgae , Soil , Biomass , Urease , Glucosylceramidase , Carbon , Arylsulfatases , Phosphoric Monoester Hydrolases , Soil Microbiology
4.
Int J Mol Sci ; 23(3)2022 Jan 30.
Article in English | MEDLINE | ID: mdl-35163551

ABSTRACT

For years, the gold standard for diagnosing Gaucher disease (GD) has been detecting reduced ß-glucocerebrosidase (GCase) activity in peripheral blood cells combined with GBA1 mutation analysis. The use of dried blood spot (DBS) specimens offers many advantages, including easy collection, the need for a small amount of blood, and simpler transportation. However, DBS has limitations for measuring GCase activity. In this paper, we recount our cross-sectional study and publish seven years of experience using DBS samples and levels of the deacylated form of glucocerebroside, glucosylsphingosine (lyso-Gb1), for GD diagnosis. Of 444 screened subjects, 99 (22.3%) were diagnosed with GD at a median (range) age of 21 (1-78) years. Lyso-Gb levels for genetically confirmed GD patients vs. subjects negative to GD diagnosis were 252 (9-1340) ng/mL and 5.4 (1.5-16) ng/mL, respectively. Patients diagnosed with GD1 and mild GBA1 variants had lower median (range) lyso-Gb1, 194 (9-1050), compared to GD1 and severe GBA1 variants, 447 (38-1340) ng/mL, and neuronopathic GD, 325 (116-1270) ng/mL (p = 0.001). Subjects with heterozygous GBA1 variants (carrier) had higher lyso-Gb1 levels, 5.8 (2.5-15.3) ng/mL, compared to wild-type GBA1, 4.9 (1.5-16), ng/mL (p = 0.001). Lyso-Gb1 levels, median (range), were 5 (2.7-10.7) in heterozygous GBA1 carriers with Parkinson's disease (PD), similar to lyso-Gb1 levels in subjects without PD. We call for a paradigm change for the diagnosis of GD based on lyso-Gb1 measurements and confirmatory GBA1 mutation analyses in DBS. Lyso-Gb1 levels could not be used to differentiate between heterozygous GBA1 carriers and wild type.


Subject(s)
Biomarkers/blood , Gaucher Disease/diagnosis , Glucosylceramidase/genetics , Psychosine/analogs & derivatives , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Cross-Sectional Studies , Early Detection of Cancer , Female , Gaucher Disease/blood , Gaucher Disease/genetics , Humans , Infant , Male , Middle Aged , Mutation , Psychosine/blood , Young Adult
5.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34622801

ABSTRACT

Current treatments for Parkinson's disease (PD) provide only symptomatic relief, with no disease-modifying therapies identified to date. Repurposing FDA-approved drugs to treat PD could significantly shorten the time needed for and reduce the costs of drug development compared with conventional approaches. We developed an efficient strategy to screen for modulators of ß-glucocerebrosidase (GCase), a lysosomal enzyme that exhibits decreased activity in patients with PD, leading to accumulation of the substrate glucosylceramide and oxidized dopamine and α-synuclein, which contribute to PD pathogenesis. Using a GCase fluorescent probe and affinity-based fluorescence polarization assay, we screened 1280 structurally diverse, bioactive, and cell-permeable FDA-approved drugs and found that the antipsychotic quetiapine bound GCase with high affinity. Moreover, quetiapine treatment of induced pluripotent stem cell-derived (iPSC-derived) dopaminergic neurons from patients carrying mutations in GBA1 or LRRK2 led to increased wild-type GCase protein levels and activity and partially lowered accumulation of oxidized dopamine, glucosylceramide, and α-synuclein. Similarly, quetiapine led to activation of wild-type GCase and reduction of α-synuclein in a GBA mutant mouse model (Gba1D409V/+ mice). Together, these results suggest that repurposing quetiapine as a modulator of GCase may be beneficial for patients with PD exhibiting decreased GCase activity.


Subject(s)
Antipsychotic Agents/pharmacology , Dopaminergic Neurons/drug effects , Glucosylceramidase/drug effects , Parkinson Disease/genetics , Parkinsonian Disorders/genetics , Quetiapine Fumarate/pharmacology , alpha-Synuclein/drug effects , Animals , Dopaminergic Neurons/metabolism , Drug Evaluation, Preclinical , Drug Repositioning , Glucosylceramidase/genetics , Glucosylceramides/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , alpha-Synuclein/metabolism
6.
J Parkinsons Dis ; 10(3): 1123-1132, 2020.
Article in English | MEDLINE | ID: mdl-32310186

ABSTRACT

BACKGROUND: Both genetic and environmental factors contribute to Parkinson's disease (PD) risk. OBJECTIVE: We investigated the potential association of several relevant variables with PD age at onset (AAO), focusing on LRRK2 p.G2019S and GBA p.N370S mutations. METHODS: Ashkenazi Jewish (AJ) PD patients, screened for LRRK2 and GBA mutations, underwent an interview regarding exposure to the following environmental and lifestyle factors: cigarette smoking, consumption of coffee, tea and alcohol, head injury and rural living. Multivariate linear regression (adjusted for sex) was used to examine the association with AAO, and models included LRRK2 p.G2019S and GBA p.N370S mutation status (carrier/non-carriers), single environmental variable and their interactions terms, as independent variables. RESULTS: 225 Israeli AJ PD patients were enrolled: 65 LRRK2 p.G2019S mutation carriers, 60 GBA p.N370S carriers and 100 non-carries of these mutations. In the dichotomized exposure/non-exposure analyses, positive LRRK2 p.G2019S status was associated with younger AAO in all models, at nominal or near significant levels (p = 0.033-0.082). Smoking was associated with older AAO (p = 0.032), and the interaction between GBA p.N370S and history of head injury was associated with younger AAO (p = 0.049), both at nominal significance. There was no indication of a consistent main effect for GBA p.N370S status or significant LRRK2 p.G2019S-environmental factor interaction. In the dose-dependent analyses, increased coffee and tea consumption levels were associated with older AAO (p = 0.001 and p = 0.002, respectively). CONCLUSIONS: Our results suggest that genetic and environmental factors may affect AAO in PD patients, but validation in additional samples is required.


Subject(s)
Gene-Environment Interaction , Glucosylceramidase/genetics , Jews , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Parkinson Disease , Adult , Age of Onset , Aged , Coffee , Drinking Behavior/physiology , Female , Heterozygote , Humans , Israel/ethnology , Jews/genetics , Jews/statistics & numerical data , Male , Middle Aged , Parkinson Disease/ethnology , Parkinson Disease/etiology , Parkinson Disease/genetics , Tea
7.
BMC Med Genet ; 21(1): 12, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31931749

ABSTRACT

BACKGROUND: Gaucher disease (GD) is a lysosomal disorder caused by biallelic pathogenic mutations in the GBA1 gene that encodes beta-glucosidase (GCase), and more rarely, by a deficiency in the GCase activator, saposin C. Clinically, GD manifests with heterogeneous multiorgan involvement mainly affecting hematological, hepatic and neurological axes. This disorder is divided into three types, based on the absence (type I) or presence and severity (types II and III) of involvement of the central nervous system. At the cellular level, deficiency of GBA1 disturbs lysosomal storage with buildup of glucocerebroside. The consequences of disturbed lysosomal metabolism on biochemical pathways that require lysosomal processing are unknown. Abnormal systemic markers of cobalamin (Cbl, B12) metabolism have been reported in patients with GD, suggesting impairments in lysosomal handling of Cbl or in its downstream utilization events. METHODS: Cultured skin fibroblasts from control humans (n = 3), from patients with GD types I (n = 1), II (n = 1) and III (n = 1) and an asymptomatic carrier of GD were examined for their GCase enzymatic activity and lysosomal compartment intactness. Control human and GD fibroblasts were cultured in growth medium with and without 500 nM hydroxocobalamin supplementation. Cellular cobalamin status was examined via determination of metabolomic markers in cell lysate (intracellular) and conditioned culture medium (extracellular). The presence of transcobalamin (TC) in whole cell lysates was examined by Western blot. RESULTS: Cultured skin fibroblasts from GD patients exhibited reduced GCase activity compared to healthy individuals and an asymptomatic carrier of GD, demonstrating a preserved disease phenotype in this cell type. The concentrations of total homocysteine (tHcy), methylmalonic acid (MMA), cysteine (Cys) and methionine (Met) in GD cells were comparable to control levels, except in one patient with GD III. The response of these metabolomic markers to supplementation with hydroxocobalamin (HOCbl) yielded variable results. The content of transcobalamin in whole cell lysates was comparable in control human and GD patients. CONCLUSIONS: Our results indicate that cobalamin transport and cellular processing pathways are overall protected from lysosomal storage damage in GD fibroblasts. Extending these studies to hepatocytes, macrophages and plasma will shed light on cell- and compartment-specific vitamin B12 metabolism in Gaucher disease.


Subject(s)
Gaucher Disease/genetics , Glucosylceramidase/genetics , Vitamin B 12/metabolism , beta-Glucosidase/genetics , Cell Culture Techniques , Female , Fibroblasts/metabolism , Gaucher Disease/metabolism , Gaucher Disease/pathology , Homocysteine/metabolism , Humans , Lysosomes/metabolism , Lysosomes/pathology , Male , Methylmalonic Acid/metabolism , Mutation , Phenotype , Saposins/genetics , Transcobalamins/metabolism
8.
Cell Death Differ ; 27(5): 1588-1603, 2020 05.
Article in English | MEDLINE | ID: mdl-31685979

ABSTRACT

Heterozygous mutations of the lysosomal enzyme glucocerebrosidase (GBA1) represent the major genetic risk for Parkinson's disease (PD), while homozygous GBA1 mutations cause Gaucher disease, a lysosomal storage disorder, which may involve severe neurodegeneration. We have previously demonstrated impaired autophagy and proteasomal degradation pathways and mitochondrial dysfunction in neurons from GBA1 knockout (gba1-/-) mice. We now show that stimulation with physiological glutamate concentrations causes pathological [Ca2+]c responses and delayed calcium deregulation, collapse of mitochondrial membrane potential and an irreversible fall in the ATP/ADP ratio. Mitochondrial Ca2+ uptake was reduced in gba1-/- cells as was expression of the mitochondrial calcium uniporter. The rate of free radical generation was increased in gba1-/- neurons. Behavior of gba1+/- neurons was similar to gba1-/- in terms of all variables, consistent with a contribution of these mechanisms to the pathogenesis of PD. These data signpost reduced bioenergetic capacity and [Ca2+]c dysregulation as mechanisms driving neurodegeneration.


Subject(s)
Calcium/metabolism , Energy Metabolism , Glucosylceramidase/deficiency , Neurons/pathology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain/pathology , Free Radicals/metabolism , Glucosylceramidase/metabolism , Glutamic Acid/toxicity , Homeostasis/drug effects , Lipid Metabolism/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Receptors, Glutamate/metabolism
9.
Int J Hematol ; 109(3): 361-365, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30456712

ABSTRACT

Gaucher disease (GD) is caused by a hereditary deficiency of glucocerebrosidase, resulting in accumulation of glucosylceramide and potentially manifesting as hepatosplenomegaly. We report the case of a 15-month-old boy with chronic neuronopathic GD. The patient had prolonged anemia despite continued iron supplementation for 3 months. White blood count (WBC), hemoglobin (Hb), platelet count, and corrected reticulocyte count were 3,300 /µL, 8.7 g/dL, 90,000 /µL, and 0.55, respectively. The patient had microcytic hypochromic anemia with mildly elevated ferritin. Physical examination revealed hepatosplenomegaly. Bone-marrow aspiration showed sheets of Gaucher cells. Glucocerebrosidase activity in monocytes was significantly lower than normal. Genetic analysis revealed a homozygous L444P mutation of GBA, and he was diagnosed with type 1 GD. Enzyme replacement treatment (ERT) consisting of imiglucerase was initiated and was effective; WBC, Hb, and platelet count gradually normalized and the hepatosplenomegaly improved. However, when the patient entered elementary school, he showed mild impaired cognitive function, and supranuclear gaze palsy occurred the same year. He was ultimately diagnosed with type 3 GD and continued ERT. Pediatric hemato-oncologists should be aware of GD, especially when patients exhibit anemia refractory to iron therapy, radiologic bone deformity, neurologic signs or symptoms, and growth retardation.


Subject(s)
Anemia, Hypochromic , Enzyme Replacement Therapy , Gaucher Disease , Glucosylceramidase/therapeutic use , Amino Acid Substitution , Anemia, Hypochromic/blood , Anemia, Hypochromic/diagnosis , Anemia, Hypochromic/drug therapy , Anemia, Hypochromic/genetics , Blood Cell Count , Bone Marrow/metabolism , Gaucher Disease/blood , Gaucher Disease/diagnosis , Gaucher Disease/drug therapy , Gaucher Disease/genetics , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Hemoglobins/metabolism , Humans , Infant , Male , Mutation, Missense
10.
Parkinsonism Relat Disord ; 59: 32-38, 2019 02.
Article in English | MEDLINE | ID: mdl-30391183

ABSTRACT

Disease modification remains a major unmet need in Parkinson's disease (PD) therapeutics. Despite multiple attempts, not a single study has yet been successful, perhaps due to our incomplete understanding of the underlying disease mechanisms. Genetic and epidemiologic studies of the last decade have substantially increased our comprehension of the etiology of PD. Once considered a pure sporadic disease, the discovery of familial mutations provided the initial paradigm shift and it is now widely accepted that PD has a substantial genetic component. These genetic discoveries have allowed the development of novel therapeutics aimed at halting or slowing the underlying disease process, rather than just ameliorating symptoms. Here, we discuss the latest advances in therapeutics based on three genetic discoveries (SNCA, LRRK2 and GBA) that are currently reaching the clinical arena and outline the challenges of therapeutic development of genetically targeted therapeutics.


Subject(s)
Biological Therapy , Glucosylceramidase , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Parkinson Disease/therapy , alpha-Synuclein , Glucosylceramidase/drug effects , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/drug effects , alpha-Synuclein/drug effects
11.
Brain Topogr ; 31(6): 1029-1036, 2018 11.
Article in English | MEDLINE | ID: mdl-29846835

ABSTRACT

Cerebral atrophy has been detected in patients with Parkinson's disease (PD) both with and without dementia, however differentiation based on genetic status has thus far not yielded robust findings. We assessed cortical thickness and subcortical volumes in a cohort of PD patients and healthy controls carriers of the G2019S mutation in the LRRK2 gene and the common GBA mutations, in an attempt to determine whether genetic status influences structural indexes. Cortical thickness and subcortical volumes were computed and compared between six groups of participants; idiopathic PD, GBA-PD, LRRK2-PD, non-manifesting non-carriers (NMNC), GBA-non-manifesting carriers (NMC) and LRRK2-NMC utilizing the FreeSurfer software program. All participants were cognitively intact based on a computerized cognitive assessment battery. Fifty-seven idiopathic PD patients, 9 LRRK2-PD, 12 GBA-PD, 49 NMNC, 41 LRRK2-NMC and 14 GBA-NMC participated in this study. Lower volumes among patients with PD compared to unaffected participants were detected in bilateral hippocampus, nucleus accumbens, caudate, thalamus, putamen and amygdala and the right pallidum (p = 0.016). PD patients demonstrated lower cortical thickness indexes in a majority of regions assessed compared with non-manifesting participants. No differences in cortical thickness and subcortical volumes were detected within each of the groups of participants based on genetic status. Mutations in the GBA and LRRK2 genes are not important determinants of cortical thickness and subcortical volumes in both patients with PD and non-manifesting participants. PD is associated with a general reduction in cortical thickness and sub-cortical atrophy even in cognitively intact patients.


Subject(s)
Brain/diagnostic imaging , Parkinson Disease/diagnostic imaging , Aged , Amygdala/diagnostic imaging , Amygdala/pathology , Biomarkers , Brain/pathology , Case-Control Studies , Caudate Nucleus/diagnostic imaging , Caudate Nucleus/pathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cohort Studies , Family , Female , Globus Pallidus/diagnostic imaging , Globus Pallidus/pathology , Glucosylceramidase/genetics , Hippocampus/diagnostic imaging , Hippocampus/pathology , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Male , Middle Aged , Mutation , Nucleus Accumbens/diagnostic imaging , Nucleus Accumbens/pathology , Organ Size , Parkinson Disease/genetics , Putamen/diagnostic imaging , Putamen/pathology , Thalamus/diagnostic imaging , Thalamus/pathology
12.
Molecules ; 23(4)2018 04 17.
Article in English | MEDLINE | ID: mdl-29673163

ABSTRACT

A series of sp²-iminosugar glycomimetics differing in the reducing or nonreducing character, the configurational pattern (d-gluco or l-ido), the architecture of the glycone skeleton, and the nature of the nonglycone substituent has been synthesized and assayed for their inhibition properties towards commercial glycosidases. On the basis of their affinity and selectivity towards GH1 ß-glucosidases, reducing and nonreducing bicyclic derivatives having a hydroxylation profile of structural complementarity with d-glucose and incorporating an N'-octyl-isourea or -isothiourea segment were selected for further evaluation of their inhibitory/chaperoning potential against human glucocerebrosidase (GCase). The 1-deoxynojirimycin (DNJ)-related nonreducing conjugates behaved as stronger GCase inhibitors than the reducing counterparts and exhibited potent chaperoning capabilities in Gaucher fibroblasts hosting the neuronopathic G188S/G183W mutation, the isothiourea derivative being indeed one of the most efficient chaperone candidates reported up to date (70% activity enhancement at 20 pM). At their optimal concentration, the four selected compounds promoted mutant GCase activity enhancements over 3-fold; yet, the inhibitor/chaperoning balance became unfavorable at much lower concentration for nonreducing as compared to reducing derivatives.


Subject(s)
Enzyme Inhibitors/therapeutic use , Gaucher Disease/drug therapy , Gaucher Disease/enzymology , Glucosylceramidase/antagonists & inhibitors , Glucosylceramidase/genetics , Imino Sugars/therapeutic use , Molecular Chaperones/therapeutic use , 1-Deoxynojirimycin/therapeutic use , Fibroblasts/drug effects , Fibroblasts/metabolism , Gaucher Disease/genetics , Glucosamine/analogs & derivatives , Glucosamine/therapeutic use , Humans , Mutation
13.
J Med Case Rep ; 12(1): 19, 2018 Jan 27.
Article in English | MEDLINE | ID: mdl-29373994

ABSTRACT

BACKGROUND: Intravenous enzyme replacement therapy is a first-line therapy for Gaucher disease type 1, and substrate reduction therapy represents an oral treatment alternative. Both enzyme replacement therapy and substrate reduction therapy are generally used as monotherapies in Gaucher disease. However, one randomized study and several case reports have described combination therapy over short time periods. CASE PRESENTATION: We report two female Gaucher disease type 1 patients of mainly Anglo-Saxon descent, where combined enzyme replacement therapy and miglustat substrate reduction therapy were administered to overcome refractory clinical symptoms. The first patient was diagnosed at age 17 and developed Gaucher disease-related bone manifestations that worsened despite starting imiglucerase enzyme replacement therapy. After switching to miglustat substrate reduction therapy, her bone symptoms improved, but she developed tremors and eventually switched back to enzyme replacement therapy. Miglustat was later recommenced in combination with ongoing enzyme replacement therapy due to continued bone pain, and her bone symptoms improved along with maintained visceral manifestations. Enzyme replacement therapy was subsequently tapered off and the patient has since been successfully maintained on miglustat. The second patient was diagnosed aged 3, and commenced imiglucerase enzyme replacement therapy aged 15. After 9 years on enzyme replacement therapy she switched to miglustat substrate reduction therapy and her core symptoms were maintained/stable for 3 years. Imiglucerase enzyme replacement therapy was later added as a boost to therapy and her symptoms were subsequently maintained over a 2.3-year period. However, miglustat was discontinued due to her relocation, necessitating an increase in enzyme replacement therapy dose. Overall, both patients benefited from combination therapy. CONCLUSION: While the majority of Gaucher disease type 1 patients will not need treatment with both substrate reduction therapy and enzyme replacement therapy, the current case reports demonstrate that judicious use of combination therapy may be of benefit in some cases.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Enzyme Inhibitors/administration & dosage , Enzyme Replacement Therapy/methods , Gaucher Disease/therapy , Glucosylceramidase/administration & dosage , 1-Deoxynojirimycin/administration & dosage , Adult , Child, Preschool , Combined Modality Therapy , Drug Therapy, Combination , Female , Hexosaminidases/blood , Humans , Thrombocytopenia/blood
15.
J Lipid Res ; 58(12): 2299-2309, 2017 12.
Article in English | MEDLINE | ID: mdl-29025868

ABSTRACT

Epidermal ß-glucocerebrosidase (GBA1), an acid ß-glucosidase normally located in lysosomes, converts (glucosyl)ceramides into ceramides, which is crucial to generate an optimal barrier function of the outermost skin layer, the stratum corneum (SC). Here we report on two developed in situ methods to localize active GBA in human epidermis: i) an optimized zymography method that is less labor intensive and visualizes enzymatic activity with higher resolution than currently reported methods using either substrate 4-methylumbelliferyl-ß-D-glucopyranoside or resorufin-ß-D-glucopyranoside; and ii) a novel technique to visualize active GBA1 molecules by their specific labeling with a fluorescent activity-based probe (ABP), MDW941. The latter method pro-ved to be more robust and sensitive, provided higher resolution microscopic images, and was less prone to sample preparation effects. Moreover, in contrast to the zymography substrates that react with various ß-glucosidases, MDW941 specifically labeled GBA1. We demonstrate that active GBA1 in the epidermis is primarily located in the extracellular lipid matrix at the interface of the viable epidermis and the lower layers of the SC. With ABP-labeling, we observed reduced GBA1 activity in 3D-cultured skin models when supplemented with the reversible inhibitor, isofagomine, irrespective of GBA expression. This inhibition affected the SC ceramide composition: MS analysis revealed an inhibitor-dependent increase in the glucosylceramide:ceramide ratio.


Subject(s)
Enzyme Assays , Fluorescent Dyes/chemistry , Glucosylceramidase/analysis , Skin/enzymology , Staining and Labeling/methods , Benzoxazines/chemistry , Boron Compounds/chemistry , Cyclohexanols/chemistry , Epoxy Compounds/chemistry , Gene Expression , Glucosides/chemistry , Glucosylceramidase/metabolism , Humans , Hymecromone/analogs & derivatives , Hymecromone/chemistry , Tissue Culture Techniques
16.
J Am Chem Soc ; 139(40): 14192-14197, 2017 10 11.
Article in English | MEDLINE | ID: mdl-28937220

ABSTRACT

Human nonlysosomal glucosylceramidase (GBA2) is one of several enzymes that controls levels of glycolipids and whose activity is linked to several human disease states. There is a major need to design or discover selective GBA2 inhibitors both as chemical tools and as potential therapeutic agents. Here, we describe the development of a fluorescence polarization activity-based protein profiling (FluoPol-ABPP) assay for the rapid identification, from a 350+ library of iminosugars, of GBA2 inhibitors. A focused library is generated based on leads from the FluoPol-ABPP screen and assessed on GBA2 selectivity offset against the other glucosylceramide metabolizing enzymes, glucosylceramide synthase (GCS), lysosomal glucosylceramidase (GBA), and the cytosolic retaining ß-glucosidase, GBA3. Our work, yielding potent and selective GBA2 inhibitors, also provides a roadmap for the development of high-throughput assays for identifying retaining glycosidase inhibitors by FluoPol-ABPP on cell extracts containing recombinant, overexpressed glycosidase as the easily accessible enzyme source.


Subject(s)
Enzyme Assays/methods , Enzyme Inhibitors/pharmacology , Fluorescence Polarization/methods , Imino Sugars/pharmacology , beta-Glucosidase/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/chemistry , Glucosylceramidase , Humans , Imino Sugars/chemistry , beta-Glucosidase/metabolism
17.
Biotechnol Prog ; 33(3): 726-736, 2017 May.
Article in English | MEDLINE | ID: mdl-28371174

ABSTRACT

OBJECTIVE: For efficient biofarming we attempted to enrich plant interstitial fluid (IF)/apoplastic fluid with targeted recombinant therapeutic protein. We employed a synthetic human Glucocerebrosidase (GCB), a model biopharmaceutical protein gene in this study. RESULTS: Twenty one Nicotiana varieties, species and hybrids were initially screened for individual IF recovery and based on the findings, we selected Nicotiana tabacum NN (S-9-6), Nicotiana tabacum nn (S-9-7) and Nicotiana benthamiana (S-6-6) as model plants for raising transgenic expressing GCB via Agrobacterium mediated transformation under the control of M24 promoter; GCB specific activity in each transgenic lines were analyzed and we observed higher concentration of recombinant GCB in IF of these transgenic lines (S-9-6, S-9-7, and S-6-6) in comparison to their concentration in crude leaf extracts. CONCLUSION: Recovery of valuable therapeutics in plant IF as shown in the present study holds great promise for promoting plant based biofarming. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:726-736, 2017.


Subject(s)
Glucosylceramidase/metabolism , Plant Extracts/metabolism , Plants, Genetically Modified/metabolism , Gene Expression Regulation, Plant/genetics , Gene Expression Regulation, Plant/physiology , Glucosylceramidase/genetics , Humans , Plant Extracts/genetics , Plant Leaves/chemistry , Plants, Genetically Modified/genetics , Promoter Regions, Genetic/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Nicotiana/metabolism
18.
Orphanet J Rare Dis ; 12(1): 23, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28166796

ABSTRACT

BACKGROUND: Gaucher disease (GD) is caused by mutations in the GBA1 gene which encodes lysosomal ß-glucocerebrosidase (GCase). In GD, partial or complete loss of GCase activity causes the accumulation of the glycolipids glucosylceramide (GlcCer) and glucosylsphingosine in the lysosomes of macrophages. In this manuscript, we investigated the effects of glycolipids accumulation on lysosomal and mitochondrial function, inflammasome activation and efferocytosis capacity in a THP-1 macrophage model of Gaucher disease. In addition, the beneficial effects of coenzyme Q10 (CoQ) supplementation on cellular alterations were evaluated. Chemically-induced Gaucher macrophages were developed by differentiateing THP-1 monocytes to macrophages by treatment with phorbol 12-myristate 13-acetate (PMA) and then inhibiting intracellular GCase with conduritol B-epoxide (CBE), a specific irreversible inhibitor of GCase activity, and supplementing the medium with exogenous GlcCer. This cell model accumulated up to 16-fold more GlcCer compared with control THP-1 cells. RESULTS: Chemically-induced Gaucher macrophages showed impaired autophagy flux associated with mitochondrial dysfunction and increased oxidative stress, inflammasome activation and impaired efferocytosis. All abnormalities were partially restored by supplementation with CoQ. CONCLUSION: These data suggest that targeting mitochondria function and oxidative stress by CoQ can ameliorate the pathological phenotype of Gaucher cells. Chemically-induced Gaucher macrophages provide cellular models that can be used to investigate disease pathogenesis and explore new therapeutics for GD.


Subject(s)
Gaucher Disease/metabolism , Macrophages/drug effects , Ubiquinone/analogs & derivatives , Glucosylceramidase , Humans , Inflammasomes , Lysosomes , Mitophagy/drug effects , Mitophagy/physiology , Reactive Oxygen Species , THP-1 Cells/drug effects , THP-1 Cells/metabolism , Ubiquinone/administration & dosage , Ubiquinone/pharmacology
19.
Orphanet J Rare Dis ; 12(1): 38, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28219443

ABSTRACT

BACKGROUND: The Disease Severity Scoring System (DS3) is a validated measure for evaluating Gaucher disease type 1 (GD1) severity. We developed a new framework, consisting of health states, transition probabilities between those states, and preferences for those states (utilities) based on the DS3 to predict long-term outcomes of patients starting treatment. We defined nine mutually exclusive (alive) health states based on three DS3 categories: mild (0 ≤ DS3 ≤ 3.5) without symptoms of bone disease; mild with bone pain, mild with severe skeletal complications (SSC) defined as lytic lesions, avascular necrosis, or fracture; moderate (3.5 < DS3 ≤ 6.5) without SSC; moderate with SSC; marked (6.5 < DS3 ≤ 9.5) without SSC; marked with SSC; severe (9.5 < DS3 ≤ 19) without SSC; and severe with SSC. Health-state transition probabilities and utilities were estimated from a longitudinal sample of patients with GD1 who started enzyme replacement therapy (the DS3 Score Study). Age dependent GD1-specific mortality was derived from published data. We used a Markov state-transition model to illustrate how to estimate time spent in each health state. RESULTS: The average predicted utilities for each health state ranged from 0.76 for mild disease with no clinical symptoms of bone disease to 0.52 with severe disease with SSC. Transition probabilities depended on disease severity (DS3 score) at treatment initiation and whether patients had undergone a total splenectomy or had an intact spleen/partial splenectomy prior to starting treatment. Patients who started treatment with intact or residual spleens spent more time in better health states than those who started treatment with total splenectomy. CONCLUSIONS: This new framework, which is based on the DS3, can be used to project the long-term outcomes of GD1 patients starting treatment. The framework could also be used to compare the long-term outcomes of different GD1 treatment options. TRIAL REGISTRATION: NCT01136304 . Registered: May 31, 2010 (retrospectively registered).


Subject(s)
Gaucher Disease/drug therapy , Gaucher Disease/pathology , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/therapeutic use , Enzyme Inhibitors/therapeutic use , Enzyme Replacement Therapy , Glucosylceramidase/therapeutic use , Humans
20.
J Med Case Rep ; 11(1): 19, 2017 Jan 20.
Article in English | MEDLINE | ID: mdl-28103924

ABSTRACT

BACKGROUND: The variants of neuronopathic Gaucher disease may be viewed as a clinical phenotypic continuum divided into acute and chronic forms. The chronic neuronopathic form of Gaucher disease is characterized by a later onset of neurological symptoms and protracted neurological and visceral involvement. The first-choice treatment for nonneuronopathic Gaucher disease is enzyme replacement therapy with recombinant analogues of the deficient human enzyme glucocerebrosidase. Enzyme replacement therapy has been shown to improve hematological and bone manifestations associated with Gaucher disease, but, as with most proteins, recombinant enzymes cannot cross the blood-brain barrier, which prevents effects on neurological manifestations. Substrate reduction therapy with miglustat (N-butyldeoxynojirimycin) inhibits glucosylceramide synthase, which catalyzes the first step in glycosphingolipid synthesis. Because miglustat can cross the blood-brain barrier, it has been suggested that, combined with enzyme replacement therapy, it might be effective in treating neurological symptoms in patients with neuronopathic Gaucher disease. CASE PRESENTATION: We report observed effects of combined enzyme replacement therapy and substrate reduction therapy in a 7-year-old Caucasian boy with neuronopathic Gaucher disease who was homozygous for L444P mutations. He had received enzyme replacement therapy from the age of 18 months, and concomitant miglustat treatment was commenced, with dosing according to body surface area uptitrated over 1 month with dietary modifications when he reached the age of 30 months. He experienced mild diarrhea after commencing miglustat therapy, which decreased in frequency/severity over time. His splenomegaly was reduced, and his hematological values and plasma angiotensin-converting enzyme activity normalized. Plasma chitotriosidase also showed substantial and sustained decreases. After 5 years of combination therapy, the patient showed no signs of neurological impairment. CONCLUSIONS: This case supports the concept that oral miglustat in combination with intravenous enzyme replacement therapy may be beneficial in preventing neurological signs in patients with chronic neuronopathic Gaucher disease. The importance of dietary modifications has also been confirmed. Further follow-up studies are needed to better define the therapeutic effect of combined treatment in this Gaucher disease subtype.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Enzyme Inhibitors/administration & dosage , Enzyme Replacement Therapy , Gaucher Disease/therapy , 1-Deoxynojirimycin/administration & dosage , Administration, Intravenous , Blood-Brain Barrier/physiopathology , Child , Chronic Disease , Combined Modality Therapy , Glucosylceramidase/deficiency , Hexosaminidases/blood , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL