Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Naunyn Schmiedebergs Arch Pharmacol ; 396(7): 1563-1569, 2023 07.
Article in English | MEDLINE | ID: mdl-36795166

ABSTRACT

Niemann-Pick type C1 (NP-C1) is a lysosomal storage disease (LSD) caused by mutations in NPC1 gene that lead to defective synthesis of the respective lysosomal transporter protein and cholesterol accumulation in late endosomes/lysosomes (LE/L) compartments, as well as glycosphingolipids GM2 and GM3 in the central nervous system (CNS). Clinical presentation varies according to the age of onset and includes visceral and neurological symptoms, such as hepatosplenomegaly and psychiatric disorders. Studies have been associating the pathophysiology of NP-C1 with oxidative damage to lipids and proteins, as well as evaluating the benefits of adjuvant therapy with antioxidants for this disease. In this work, we evaluated the DNA damage in fibroblasts culture from patients with NP-C1 treated with miglustat, as well as the in vitro effect of the antioxidant compounds N-acetylcysteine (NAC) and Coenzyme Q10 (CoQ10), using the alkaline comet assay. Our preliminary results demonstrate that NP-C1 patients have increased DNA damage compared to healthy individuals and that the treatments with antioxidants can mitigate it. DNA damage may be due to an increase in reactive species since it has been described that NP-C1 patients have increased peripheral markers of damage to other biomolecules. Our study suggests that NP-C1 patients could benefit from the use of adjuvant therapy with NAC and CoQ10, which should be better evaluated in a future clinical trial.


Subject(s)
Niemann-Pick Disease, Type C , Humans , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/metabolism , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , DNA Damage
2.
Biofactors ; 47(4): 551-569, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33878238

ABSTRACT

Coenzyme Q10 (CoQ10 ) deficiency is a rare disease characterized by a decreased accumulation of CoQ10 in cell membranes. Considering that CoQ10 synthesis and most of its functions are carried out in mitochondria, CoQ10 deficiency cases are usually considered a mitochondrial disease. A relevant feature of CoQ10 deficiency is that it is the only mitochondrial disease with a successful therapy available, the CoQ10 supplementation. Defects in components of the synthesis machinery caused by mutations in COQ genes generate the primary deficiency of CoQ10 . Mutations in genes that are not directly related to the synthesis machinery cause secondary deficiency. Cases of CoQ10 deficiency without genetic origin are also considered a secondary deficiency. Both types of deficiency can lead to similar clinical manifestations, but the knowledge about primary deficiency is deeper than secondary. However, secondary deficiency cases may be underestimated since many of their clinical manifestations are shared with other pathologies. This review shows the current state of secondary CoQ10 deficiency, which could be even more relevant than primary deficiency for clinical activity. The analysis covers the fundamental features of CoQ10 deficiency, which are necessary to understand the biological and clinical differences between primary and secondary CoQ10 deficiencies. Further, a more in-depth analysis of CoQ10 secondary deficiency was undertaken to consider its origins, introduce a new way of classification, and include aging as a form of secondary deficiency.


Subject(s)
Aging/genetics , Alkyl and Aryl Transferases/genetics , Ataxia/genetics , GTP Phosphohydrolases/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Muscle Weakness/genetics , Niemann-Pick Disease, Type C/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Aging/metabolism , Alkyl and Aryl Transferases/metabolism , Animals , Ataxia/metabolism , Ataxia/pathology , Energy Metabolism/genetics , GTP Phosphohydrolases/metabolism , Gene Expression Regulation , Humans , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/metabolism , Muscle Weakness/metabolism , Muscle Weakness/pathology , Mutation , Niemann-Pick C1 Protein/genetics , Niemann-Pick C1 Protein/metabolism , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Signal Transduction , Ubiquinone/genetics , Ubiquinone/metabolism
3.
Int J Mol Sci ; 21(12)2020 Jun 24.
Article in English | MEDLINE | ID: mdl-32599915

ABSTRACT

Niemann-Pick type C1 (NPC1) is a lysosomal storage disorder, inherited as an autosomal-recessive trait. Mutations in the Npc1 gene result in malfunction of the NPC1 protein, leading to an accumulation of unesterified cholesterol and glycosphingolipids. Beside visceral symptoms like hepatosplenomegaly, severe neurological symptoms such as ataxia occur. Here, we analyzed the sphingosine-1-phosphate (S1P)/S1P receptor (S1PR) axis in different brain regions of Npc1-/- mice and evaluated specific effects of treatment with 2-hydroxypropyl-ß-cyclodextrin (HPßCD) together with the iminosugar miglustat. Using high-performance thin-layer chromatography (HPTLC), mass spectrometry, quantitative real-time PCR (qRT-PCR) and western blot analyses, we studied lipid metabolism in an NPC1 mouse model and human skin fibroblasts. Lipid analyses showed disrupted S1P metabolism in Npc1-/- mice in all brain regions, together with distinct changes in S1pr3/S1PR3 and S1pr5/S1PR5 expression. Brains of Npc1-/- mice showed only weak treatment effects. However, side effects of the treatment were observed in Npc1+/+ mice. The S1P/S1PR axis seems to be involved in NPC1 pathology, showing only weak treatment effects in mouse brain. S1pr expression appears to be affected in human fibroblasts, induced pluripotent stem cells (iPSCs)-derived neural progenitor and neuronal differentiated cells. Nevertheless, treatment-induced side effects make examination of further treatment strategies indispensable.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Brain/drug effects , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/physiology , Lysophospholipids/metabolism , Mutation , Niemann-Pick Disease, Type C/drug therapy , Sphingosine/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , Adult , Animals , Brain/metabolism , Brain/pathology , Glycoside Hydrolase Inhibitors/pharmacology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mice , Mice, Knockout , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Sphingosine/metabolism , Young Adult
4.
J Lipid Res ; 61(6): 830-839, 2020 06.
Article in English | MEDLINE | ID: mdl-32291331

ABSTRACT

Niemann-Pick type C (NPC)1 disease is a rare genetic condition in which the function of the lysosomal cholesterol transporter NPC1 protein is impaired. Consequently, sphingolipids and cholesterol accumulate in lysosomes of all tissues, triggering a cascade of pathological events that culminate in severe systemic and neurological symptoms. Lysosomal cholesterol accumulation is also a key factor in the development of atherosclerosis and NASH. In these two metabolic diseases, the administration of plant stanol esters has been shown to ameliorate cellular cholesterol accumulation and inflammation. Given the overlap of pathological mechanisms among atherosclerosis, NASH, and NPC1 disease, we sought to investigate whether dietary supplementation with plant stanol esters improves the peripheral features of NPC1 disease. To this end, we used an NPC1 murine model featuring a Npc1-null allele (Npc1nih ), creating a dysfunctional NPC1 protein. Npc1nih mice were fed a 2% or 6% plant stanol ester-enriched diet over the course of 5 weeks. During this period, hepatic and blood lipid and inflammatory profiles were assessed. Npc1nih mice fed the plant stanol-enriched diet exhibited lower hepatic cholesterol accumulation, damage, and inflammation than regular chow-fed Npc1nih mice. Moreover, plant stanol consumption shifted circulating T-cells and monocytes in particular toward an anti-inflammatory profile. Overall, these effects were stronger following dietary supplementation with 6% stanols, suggesting a dose-dependent effect. The findings of our study highlight the potential use of plant stanols as an affordable complementary means to ameliorate disorders in hepatic and blood lipid metabolism and reduce inflammation in NPC1 disease.


Subject(s)
Dietary Supplements , Niemann-Pick Disease, Type C/drug therapy , Sitosterols/pharmacology , Animals , Cholesterol/metabolism , Disease Models, Animal , Liver/drug effects , Liver/metabolism , Male , Mice , Niemann-Pick Disease, Type C/metabolism , Sitosterols/therapeutic use , Sphingolipids/metabolism
5.
Mol Ther ; 27(8): 1507-1526, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31138511

ABSTRACT

Gliosis in Niemann-Pick type C (NP-C) disease is characterized by marked changes in microglia and astrocytes. However, the gliosis onset and progression in NP-C has not been systematically studied, nor has the mechanism underlying this finding. Here, we found early gliosis in the subventricular zone (SVZ) of NP-C mice. Neural progenitor damage by Npc1 mutation suppressed vascular endothelial growth factor (VEGF) expression and further induced microglia activation followed by astrogliosis. Interestingly, excessive astrogliosis in the SVZ induced neural progenitor retention and/or migration into thalamus via astrocyte-derived VEGF, resulting in acceleration of thalamic and cortical gliosis through thalamo-cortical pathways. Transplantation of VEGF-overexpressing neural stem cells into the SVZ improved whole-brain pathology of NP-C mice. Overall, our data provide a new pathological perspective on NP-C neural pathology, revealing abnormalities in the subventricular-thalamo-cortical circuit of NP-C mouse brain and highlighting the importance of the SVZ microenvironment as a therapeutic target for NP-C disease.


Subject(s)
Cerebral Cortex/metabolism , Lateral Ventricles/metabolism , Niemann-Pick Disease, Type C/metabolism , Signal Transduction , Thalamus/metabolism , Animals , Astrocytes/metabolism , Biomarkers , Cell Movement , Disease Models, Animal , Gliosis/etiology , Gliosis/metabolism , Gliosis/pathology , Mice , Microglia/metabolism , Neural Stem Cells/metabolism , Niemann-Pick Disease, Type C/etiology , Niemann-Pick Disease, Type C/pathology , Niemann-Pick Disease, Type C/therapy , Vascular Endothelial Growth Factor A/metabolism
6.
Int J Mol Sci ; 19(4)2018 Mar 24.
Article in English | MEDLINE | ID: mdl-29587349

ABSTRACT

Niemann-Pick-disease type C1 (NPC1) is an autosomal-recessive cholesterol-storage disorder. Besides other symptoms, NPC1 patients develop liver dysfunction and hepatosplenomegaly. The mechanisms of hepatomegaly and alterations of lipid metabolism-related genes in NPC1 disease are still poorly understood. Here, we used an NPC1 mouse model to study an additive hepatoprotective effect of a combination of 2-hydroxypropyl-ß-cyclodextrin (HPßCD), miglustat and allopregnanolone (combination therapy) with the previously established monotherapy using HPßCD. We examined transgene effects as well as treatment effects on liver morphology and hepatic lipid metabolism, focusing on hepatic cholesterol transporter genes. Livers of Npc1-/- mice showed hepatic cholesterol sequestration with consecutive liver injury, an increase of lipogenetic gene expression, e.g., HMG-CoA, a decrease of lipolytic gene expression, e.g., pparα and acox1, and a decrease of lipid transporter gene expression, e.g., acat1, abca1 and fatp2. Both, combination therapy and monotherapy, led to a reduction of hepatic lipids and an amelioration of NPC1 liver disease symptoms. Monotherapy effects were related to pparα- and acox1-associated lipolysis/ß-oxidation and to fatp2-induced fatty acid transport, whereas the combination therapy additionally increased the cholesterol transport via abca1 and apoE. However, HPßCD monotherapy additionally increased cholesterol synthesis as indicated by a marked increase of the HMG-CoA and srebp-2 mRNA expression, probably as a result of increased hepatocellular proliferation.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , 2-Hydroxypropyl-beta-cyclodextrin/administration & dosage , Hepatomegaly/drug therapy , Hepatomegaly/etiology , Liver/pathology , Niemann-Pick Disease, Type C/complications , Niemann-Pick Disease, Type C/drug therapy , Pregnanolone/administration & dosage , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/therapeutic use , 2-Hydroxypropyl-beta-cyclodextrin/therapeutic use , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Cholesterol/metabolism , Disease Models, Animal , Drug Therapy, Combination , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Mice, Knockout , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Pregnanolone/therapeutic use , Proteins/genetics , Proteins/metabolism
7.
Int J Mol Sci ; 17(11)2016 Nov 09.
Article in English | MEDLINE | ID: mdl-27834854

ABSTRACT

Niemann-Pick Type C1 (NPC1) is an autosomal recessive inherited disorder characterized by accumulation of cholesterol and glycosphingolipids. Previously, we demonstrated that BALB/c-npc1nihNpc1-/- mice treated with miglustat, cyclodextrin and allopregnanolone generally performed better than untreated Npc1-/- animals. Unexpectedly, they also seemed to accomplish motor tests better than their sham-treated wild-type littermates. However, combination-treated mutant mice displayed worse cognition performance compared to sham-treated ones. To evaluate effects of these drugs in healthy BALB/c mice, we here analyzed pharmacologic effects on motor and cognitive behavior of wild-type mice. For combination treatment mice were injected with allopregnanolone/cyclodextrin weekly, starting at P7. Miglustat injections were performed daily from P10 till P23. Starting at P23, miglustat was embedded in the chow. Other mice were treated with miglustat only, or sham-treated. The battery of behavioral tests consisted of accelerod, Morris water maze, elevated plus maze, open field and hot-plate tests. Motor capabilities and spontaneous motor behavior were unaltered in both drug-treated groups. Miglustat-treated wild-type mice displayed impaired spatial learning compared to sham- and combination-treated mice. Both combination- and miglustat-treated mice showed enhanced anxiety in the elevated plus maze compared to sham-treated mice. Additionally, combination treatment as well as miglustat alone significantly reduced brain weight, whereas only combination treatment reduced body weight significantly. Our results suggest that allopregnanolone/cyclodextrin ameliorate most side effects of miglustat in wild-type mice.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Behavior, Animal/drug effects , Cognition/drug effects , Cyclodextrins/pharmacology , Hypolipidemic Agents/pharmacology , Pregnanolone/pharmacology , 1-Deoxynojirimycin/pharmacology , Animals , Body Weight/drug effects , Brain/drug effects , Brain/physiology , Disease Models, Animal , Drug Administration Schedule , Drug Combinations , Male , Maze Learning/drug effects , Mice , Mice, Inbred BALB C , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/physiopathology , Organ Size/drug effects
8.
Int J Mol Sci ; 17(7)2016 Jul 04.
Article in English | MEDLINE | ID: mdl-27384562

ABSTRACT

Lysosomal storage diseases (LSDs) are a group of rare, life-threatening genetic disorders, usually caused by a dysfunction in one of the many enzymes responsible for intralysosomal digestion. Even though no cure is available for any LSD, a few treatment strategies do exist. Traditionally, efforts have been mainly targeting the functional loss of the enzyme, by injection of a recombinant formulation, in a process called enzyme replacement therapy (ERT), with no impact on neuropathology. This ineffectiveness, together with its high cost and lifelong dependence is amongst the main reasons why additional therapeutic approaches are being (and have to be) investigated: chaperone therapy; gene enhancement; gene therapy; and, alternatively, substrate reduction therapy (SRT), whose aim is to prevent storage not by correcting the original enzymatic defect but, instead, by decreasing the levels of biosynthesis of the accumulating substrate(s). Here we review the concept of substrate reduction, highlighting the major breakthroughs in the field and discussing the future of SRT, not only as a monotherapy but also, especially, as complementary approach for LSDs.


Subject(s)
Lysosomal Storage Diseases/therapy , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/therapeutic use , Enzyme Replacement Therapy , Gaucher Disease/metabolism , Gaucher Disease/pathology , Gaucher Disease/therapy , Genistein/therapeutic use , Humans , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Mucopolysaccharidoses/metabolism , Mucopolysaccharidoses/pathology , Mucopolysaccharidoses/therapy , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Niemann-Pick Disease, Type C/therapy
9.
J Neurol ; 263(5): 927-936, 2016 May.
Article in English | MEDLINE | ID: mdl-26984608

ABSTRACT

Niemann-Pick disease type C (NP-C) is a fatal progressive neurolipidosis involving neuronal storage of cholesterol and gangliosides. Miglustat, an inhibitor of glycosphingolipid synthesis, has been approved to treat neurological manifestations in adults and children with NP-C. This open-label observational study in adults with confirmed NP-C evaluated the efficacy of miglustat (200 mg t.i.d.) based on composite functional disability (CFD) scores and brain proton magnetic resonance spectroscopy (H-MRS) measurement of choline (Cho)/N-acetyl aspartate (NAA) ratio in the centrum ovale. Overall, 16 patients were included and received miglustat for a mean period of 30.6 months: 12 continued on miglustat throughout follow up, and 4 discontinued miglustat because of adverse effects (n = 2) or perceived lack of efficacy (n = 2). In the 'continued' subgroup, the mean (SD) annual progression of CFD scores decreased from 0.75 (0.94) before treatment to 0.29 (1.29) during the period between miglustat initiation and last follow-up. In the discontinued subgroup, CFD progression increased from 0.48 (0.44) pre-treatment to 1.49 (1.31) at last follow up (off treatment). Mean (SD) Cho/NAA ratio [normal level 0.48 (0.076)] decreased during miglustat treatment in the continued subgroup: 0.64 (0.12) at baseline (miglustat initiation), 0.59 (0.17) at 12-month follow up, and 0.48 (0.09) at 24-month follow up. Cho/NAA ratio remained relatively stable in the discontinued subgroup: 0.57 (0.15), 0.53 (0.04) and 0.55 (0.09), respectively. In conclusion, H-MRS Cho/NAA ratio might serve as an objective, quantitative neurological marker of brain dysfunction in NP-C, allowing longitudinal analysis of the therapeutic effect of miglustat.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Brain/drug effects , Brain/diagnostic imaging , Enzyme Inhibitors/therapeutic use , Niemann-Pick Disease, Type C/diagnostic imaging , Niemann-Pick Disease, Type C/drug therapy , 1-Deoxynojirimycin/adverse effects , 1-Deoxynojirimycin/therapeutic use , Adolescent , Adult , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Brain/metabolism , Choline/metabolism , Disability Evaluation , Disease Progression , Enzyme Inhibitors/adverse effects , Female , Follow-Up Studies , Humans , Linear Models , Longitudinal Studies , Male , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/metabolism , Patient Dropouts , Treatment Outcome , Young Adult
10.
J Vet Med Sci ; 77(6): 693-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25715651

ABSTRACT

Niemann-Pick type C disease (NP-C) is a rare and ultimately fatal lysosomal storage disorder with variable neurologic symptoms. Loss of neuronal function and neuronal cell death occur in the NP-C brain, similar to the findings for other neurodegenerative diseases. Targeting of neuronal cells in the brain therefore represents a potential clinical intervention strategy to reduce the rate of disease progression and improve the quality of life. We previously reported that bone marrow stem cells show a neurogenic effect through CCL2 (also known as monocyte chemoattractant protein-1, MCP-1) secretion in the brains of NP-C mice. However, the direct effect of CCL2 on neurogenesis has not been ascertained. Here, to define neurogenic effects of CCL2 in NP-C, we applied human recombinant CCL2 to neural stem cells (NSCs) derived from NP-C mice. CCL2-treated NSCs showed significantly increased capacity for self-renewal, proliferation and neuronal differentiation. Similar results were observed in the subventricular zone of NP-C mice after CCL2 treatment. Furthermore, infusion of CCL2 into the NP-C mouse brain resulted in reduction of neuroinflammation. Taken together, our results demonstrate that CCL2 is a potential new therapeutic agent for NP-C.


Subject(s)
Chemokine CCL2/pharmacology , Drug Evaluation, Preclinical/methods , Neural Stem Cells/physiology , Neurogenesis/drug effects , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/metabolism , Analysis of Variance , Animals , Bromodeoxyuridine , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chemokine CCL2/metabolism , DNA Primers/genetics , Immunohistochemistry , Mice , Mice, Inbred BALB C , Neural Stem Cells/drug effects , Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
11.
Biochemistry (Mosc) ; 78(7): 721-5, 2013 Jul.
Article in English | MEDLINE | ID: mdl-24010835

ABSTRACT

The lysosomal storage disorders (LSD) represent a heterogeneous group of inherited diseases characterized by the accumulation of non-metabolized macromolecules (by-products of cellular turnover) in different tissues and organs. LSDs primarily develop as a consequence of a deficiency in a lysosomal hydrolase or its co-factor. The majority of these enzymes are glycosidases and sulfatases, which in normal conditions participate in degradation of glycoconjugates: glycoproteins, glycosaminoproteoglycans, and glycolipids. Significant insights have been gained from studies of animal models, both in understanding mechanisms of disease and in establishing proof of therapeutic concept. These studies have led to the introduction of therapy for certain LSD subtypes, primarily by enzyme replacement or substrate reduction therapy. Animal models have been useful in elucidating molecular changes, particularly prior to onset of symptoms. On the other hand, it should be noted certain animal (mouse) models may have the underlying biochemical defect, but not show the course of disease observed in human patients. There is interest in examining therapeutic options in the larger spontaneous animal models that may more closely mimic the brain size and pathology of humans. This review will highlight lessons learned from studies of animal models of disease, drawing primarily from publications in 2011-2012.


Subject(s)
Lysosomal Storage Diseases/metabolism , Aminopeptidases/genetics , Aminopeptidases/metabolism , Aminopeptidases/therapeutic use , Animals , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/metabolism , Glycogen Storage Disease Type II/pathology , Humans , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/pathology , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Serine Proteases/genetics , Serine Proteases/metabolism , Serine Proteases/therapeutic use , Tripeptidyl-Peptidase 1 , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism , alpha-Glucosidases/therapeutic use
12.
Neurol Neurochir Pol ; 47(1): 86-9, 2013.
Article in English | MEDLINE | ID: mdl-23487299

ABSTRACT

Niemann-Pick disease type C is a rare hereditary disorder caused by mutation-disrupted metabolism of cholesterol and low-density lipoprotein (LDL). In most patients, symptoms begin in childhood with severe clinical progression. We present a patient with heterozygote mutations 3001A>G and 3019C>G with late onset of the disease and positive response to treatment with miglustat. Behaviour and educational problems in childhood were probably related to the disease diagnosed later.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Enzyme Inhibitors/administration & dosage , Niemann-Pick Disease, Type C/diagnosis , Niemann-Pick Disease, Type C/drug therapy , Phenotype , 1-Deoxynojirimycin/administration & dosage , Humans , Male , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/metabolism , Young Adult
13.
J Nutr ; 142(11): 1935-42, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22990467

ABSTRACT

The Niemann-Pick C1 (NPC1) gene is associated with human obesity. Mouse models with decreased Npc1 gene dosage are susceptible to weight gain when fed a high-fat diet, but not a low-fat diet, consistent with an Npc1 gene-diet interaction. The objectives of this study were to define regulation of the Npc1 gene and to investigate the Npc1 gene-diet interaction responsible for weight gain. The experimental design involved feeding C57BL/6J male mice a low-fat diet (with 0.00, 0.10, or 1.00% cholesterol) or a high-fat diet (with 0.02% cholesterol) until 30 wk to determine regulation of the Npc1 gene in liver. The key results showed that the Npc1 gene was downregulated by dietary fatty acids (54%, P = 0.022), but not by dietary cholesterol, through feedback inhibition of the sterol regulatory element-binding protein (SREBP) pathway. However, the dietary fatty acids secondarily increased liver cholesterol, which also inhibits the SREBP pathway. Similarly, the Npc1 gene was downregulated in peritoneal fibroblasts isolated from C57BL/6J weanling male mice not exposed to the experimental diets and incubated in media supplemented with purified oleic acid (37%, P = 0.038) but not in media supplemented with purified cholesterol. These results are important because they suggest a novel mechanism for the interaction of fatty acids with the Npc1 gene to influence energy balance and to promote weight gain. Moreover, the responsiveness of the Npc1 gene to fatty acids is consistent with studies that suggest that the encoded NPC1 protein has a physiologic role in regulating both cholesterol and fatty acid metabolism.


Subject(s)
Cholesterol, Dietary/pharmacology , Dietary Fats/pharmacology , Down-Regulation/drug effects , Liver/drug effects , Proteins/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Body Weight , Cholesterol/pharmacology , Diet , Fibroblasts/cytology , Fibroblasts/drug effects , Intracellular Signaling Peptides and Proteins , Lipid Metabolism , Liver/anatomy & histology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/metabolism , Oleic Acid/pharmacology , Organ Size , Peritoneum/cytology , Proteins/genetics , RNA, Messenger/isolation & purification
14.
PLoS One ; 6(12): e28418, 2011.
Article in English | MEDLINE | ID: mdl-22163015

ABSTRACT

BACKGROUND: Niemann Pick disease type C1 is a neurodegenerative disease caused by mutations in the NPC1 gene, which result in accumulation of unesterified cholesterol and glycosphingolipids in the endosomal-lysosomal system as well as limiting membranes. We have previously shown the corneal involvement in NPC1 pathology in form of intracellular inclusions in epithelial cells and keratocytes. The purpose of the present study was to clarify if these inclusions regress during combined substrate reduction- and by-product therapy (SRT and BPT). METHODOLOGY/PRINCIPAL FINDINGS: Starting at postnatal day 7 (P7) and thereafter, NPC1 knock-out mice (NPC1(-/-)) and wild type controls (NPC1(+/+)) were injected with cyclodextrin/allopregnanolone weekly. Additionally, a daily miglustat injection started at P10 until P23. Starting at P23 the mice were fed powdered chow with daily addition of miglustat. The sham group was injected with 0.9% NaCl at P7, thereafter daily starting at P10 until P23, and fed powdered chow starting at P23. For corneal examination, in vivo confocal laser-scanning microscopy (CLSM) was performed one day before experiment was terminated. Excised corneas were harvested for lipid analysis (HPLC/MS) and electron microscopy. In vivo CLSM demonstrated a regression of hyperreflective inclusions in all treated NPC1(-/-)mice. The findings varied between individual mice, demonstrating a regression, ranging from complete absence to pronounced depositions. The reflectivity of inclusions, however, was significantly lower when compared to untreated and sham-injected NPC1(-/-) mice. These confocal findings were confirmed by lipid analysis and electron microscopy. Another important CLSM finding revealed a distinct increase of mature dendritic cell number in corneas of all treated mice (NPC1(-/-) and NPC1(+/+)), including sham-treated ones. CONCLUSIONS/SIGNIFICANCE: The combined substrate reduction- and by-product therapy revealed beneficial effects on the cornea. In vivo CLSM is a non-invasive tool to monitor disease progression and treatment effects in NPC1 disorder.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Cornea/drug effects , Cyclodextrins/administration & dosage , Niemann-Pick Disease, Type C/metabolism , Pregnanolone/administration & dosage , 1-Deoxynojirimycin/administration & dosage , Anesthetics/administration & dosage , Animals , Chromatography, High Pressure Liquid/methods , Chromatography, Liquid/methods , Cornea/metabolism , Cornea/pathology , Disease Progression , Drug Therapy, Combination/methods , Enzyme Inhibitors/administration & dosage , Genotype , Intracellular Signaling Peptides and Proteins , Lysosomes/metabolism , Mass Spectrometry/methods , Mice , Mice, Knockout , Microscopy, Confocal/methods , Models, Biological , Mutation , Niemann-Pick C1 Protein , Proteins/genetics
15.
Metab Brain Dis ; 26(4): 299-306, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21938520

ABSTRACT

Niemann-Pick type C 1 (NPC1) disease is an autosomal recessive cholesterol transport defect resulting in a neurodegenerative process in patients mainly at an early age, although some patients may start with manifestation in adult. Since loss of myelin is considered as a main pathogenetic factor, the precise mechanism inducing dysmylination in NPC1 disease is still unclear. In the present study, a quantitative evaluation on the myelin protein and its regulatory factors of oligodendrocytes, such as SRY-related HMG-box 10 (Sox10), Yin Yang 1 factor (YY1) and myelin gene regulatory factor (MRF), in different parts of the brain and spinal cord was performed in NPC1-mutant mice. The results showed that NPC1 protein was expressed in oligodendrocytes and the amount of myelin protein was generally decreased in all parts of the brain and spinal cord in NPC1-mutant mice. Compared to wild type, the amount of Sox10 and YY1 was not different in NPC1-mutant mice, but MRF was significantly decreased, suggesting a possible mechanism perturbing differentiation of oligodendrocytes and the myelination process in the NPC1-mutant mouse.


Subject(s)
Myelin Sheath , Nerve Degeneration , Niemann-Pick Disease, Type C , Oligodendroglia/metabolism , Proteins/metabolism , Transcription Factors/metabolism , Animals , Brain/metabolism , Brain/pathology , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Myelin Sheath/genetics , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Proteins/genetics , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Transcription Factors/genetics , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
16.
Curr Pharm Biotechnol ; 12(6): 897-901, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21235443

ABSTRACT

Niemann-Pick disease type C is an autosomal recessive disorder caused by mutations in either one of the two genes, NPC1 or NPC2, which encode proteins involved in the regulation of normal transport and/or processing of free cholesterol. Several types of lipids including free cholesterol (unesterified), sphingosine, sphingomyelin, phospholipids and glycosphingolipids (glucosylceramide and gangliosides GM2 and GM3) are accumulated in lysosomes and late endosomes of cells, with pronounced concentrations in the liver and the spleen. The key laboratory diagnostic test for NP-C is filliping staining of cultured skin fibroblasts from the patient, to demonstrate free cholesterol accumulation in lysosomes secondary to impaired intracellular cholesterol transport. The symptomatology and rate of disease progression are strongly influenced by age at disease onset and different clinical forms have been described on this basis: Perinatal, Early-infantile (EI), late-infantile (LI), juvenile and adult forms. Clinical symptoms include progressive neurological deterioration and visceral organomegaly. Nowadays there is no fully effective treatment, only supportive measures for relief of specific manifestations of the disease. The intervention to slow disease progression is the most promising therapy. A number of experimental disease - specific therapies, based on the molecular pathology of NP-C, have been tested in cell culture and animal models including neurosteroids, cholesterol - binding agents, curcumin and Miglustat. This paper summarizes the recent developments that have been investigated for the treatment in patients and animal models with NPC. Current therapeutic approaches have been classified based on the targeting of cellular function, the anti - apoptotic cellular mechanisms and the stem cells therapy.


Subject(s)
Niemann-Pick Disease, Type C/therapy , Age Factors , Animals , Cholesterol/metabolism , Humans , Molecular Targeted Therapy/methods , Mutation , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/metabolism
17.
PLoS One ; 5(9): e12941, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20886067

ABSTRACT

BACKGROUND: Niemann-Pick type C (NPC) disease is a lysosomal storage disease characterized by the accumulation of cholesterol and glycosphingolipids. The majority of NPC patients die in their teen years due to progressive neurodegeneration; however, half of NPC patients also suffer from cholestasis, prolonged jaundice, and hepatosplenomegaly. We previously showed that a key mediator of NPC liver disease is tumor necrosis factor (TNF) α, which is involved in both proinflammatory and apoptotic signaling cascades. In this study, we tested the hypothesis that blocking TNF action with an anti-TNF monoclonal antibody (CNTO5048) will slow the progression of NPC liver disease. METHODOLOGY/PRINCIPAL FINDINGS: Treatment of wild-type C57BL/6 mice with NPC1-specific antisense oligonucleotides led to knockdown of NPC1 protein expression in the liver. This caused classical symptoms of NPC liver disease, including hepatic cholesterol accumulation, hepatomegaly, elevated serum liver enzymes, and lipid laden macrophage accumulation. In addition, there was a significant increase in the number of apoptotic cells and a proliferation of stellate cells. Concurrent treatment of NPC1 knockdown mice with anti-TNF had no effect on the primary lipid storage or accumulation of lipid-laden macrophages. However, anti-TNF treatment slightly blunted the increase in hepatic apoptosis and stellate cell activation that was seen with NPC1 knockdown. CONCLUSIONS/SIGNIFICANCE: Current therapeutic options for NPC disease are limited. Our results provide proof of principle that pharmacologically blocking the TNF-α inflammatory cascade can slightly reduce certain markers of NPC disease. Small molecule inhibitors of TNF that penetrate tissues and cross the blood-brain barrier may prove even more beneficial.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Disease Models, Animal , Mice , Niemann-Pick Disease, Type C/drug therapy , Tumor Necrosis Factor-alpha/immunology , Animals , Apoptosis/drug effects , Cholesterol/metabolism , Drug Evaluation, Preclinical , Female , Humans , Intracellular Signaling Peptides and Proteins , Lipid Metabolism/drug effects , Liver/cytology , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/metabolism , Proteins/genetics , Proteins/metabolism
18.
Assay Drug Dev Technol ; 8(3): 295-320, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20469965

ABSTRACT

Niemann-Pick disease type C (NPC) is an inherited lipid storage disorder characterized by a defect in intracellular trafficking of exogenous cholesterol and glycosphingolipids. A goal for therapeutic treatment of NPC is to decrease/normalize cholesterol accumulation. We developed a functional genomics-based assay, combining high-throughput RNA interference (HT-RNAi) screening with high-content fluorescence imaging to identify specific genes in NPC cells that will result in more normal cholesterol levels in the diseased cells. Conditions for siRNA tranfections were optimized for 2 NPC fibroblast cell lines (GM03123, GM18453) and a normal fibroblast cell line (GM05659). RNAi screening was done using a focused-set siRNA library targeting 40 cholesterol trafficking-associated genes, knowledge mined from the existing literature on NPC disease, and/or their association with NPC1/NPC2 genes. We utilized filipin staining as a measure of cholesterol accumulation in fixed NPC cells. Data analysis of these screens confirmed several genes including LDLR and RAB9A that reduced cholesterol content in NPC cells. Nine genes were validated using filipin staining to detect unesterified cholesterol as well as cholesteryl BODIPY esters to study lipid trafficking. Gene silencing was also confirmed using qRT-PCR. Our results show that this technology can be applied to larger screens to identify genes responsible for lipid accumulation and/or trafficking in NPC disease, which could be instrumental in developing innovative therapies for individuals afflicted with NPC disease.


Subject(s)
Cholesterol/metabolism , Niemann-Pick Disease, Type C/metabolism , RNA Interference/physiology , Boron Compounds/metabolism , Cell Line , Cholesterol Esters/metabolism , Data Interpretation, Statistical , Drug Evaluation, Preclinical , Fibroblasts/metabolism , Fluorescent Dyes , Genomics , Humans , Image Processing, Computer-Assisted , Niemann-Pick Disease, Type C/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Software
19.
J Biol Chem ; 283(41): 27871-27880, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18644787

ABSTRACT

Most cell types acquire cholesterol by endocytosis of circulating low density lipoprotein, but little is known about the mechanisms of intra-endosomal cholesterol transport and about the primary cause of its aberrant accumulation in the cholesterol storage disorder Niemann-Pick type C (NPC). Here we report that lysobisphosphatidic acid (LBPA), an unconventional phospholipid that is only detected in late endosomes, regulates endosomal cholesterol levels under the control of Alix/AlP1, which is an LBPA-interacting protein involved in sorting into multivesicular endosomes. We find that Alix down-expression decreases both LBPA levels and the lumenal vesicle content of late endosomes. Cellular cholesterol levels are also decreased, presumably because the storage capacity of endosomes is affected and thus cholesterol clearance accelerated. Both lumenal membranes and cholesterol can be restored in Alix knockdown cells by exogenously added LBPA. Conversely, we also find that LBPA becomes limiting upon pathological cholesterol accumulation in NPC cells, because the addition of exogenous LBPA, but not of LBPA isoforms or analogues, partially reverts the NPC phenotype. We conclude that LBPA controls the cholesterol capacity of endosomes.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cholesterol/metabolism , Endocytosis , Endosomes/metabolism , Lysophospholipids/metabolism , Monoglycerides/metabolism , Niemann-Pick Disease, Type C/metabolism , Animals , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/genetics , Cholesterol/genetics , Cricetinae , Endocytosis/drug effects , Endocytosis/genetics , Endosomal Sorting Complexes Required for Transport , Endosomes/genetics , Endosomes/ultrastructure , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HeLa Cells , Humans , Lysophospholipids/pharmacology , Monoglycerides/pharmacology , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology
SELECTION OF CITATIONS
SEARCH DETAIL