Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-37956788

RESUMEN

Acid sphingomyelinase deficiency is a neurodegenerative lysosomal storage disorder caused by mutations in the sphingomyelin-degrading enzyme acid sphingomyelinase (ASM) gene. Upregulated neuroinflammation has been well-characterized in an ASM knockout mouse model of acid sphingomyelinase deficiency disease, but lipid mediator pathways involved in 'mediating' inflammation and inflammation-resolution have yet to be characterized. In this study, we 1) measured free (bioactive) and esterified (inactive) lipid mediators involved in inflammation and inflammation resolution in cerebellum and neuronal cultures of ASM knockout (ASMko) mice and wildtype (WT) controls, and 2) quantified the esterification of labeled pro-resolving free d11-14(15)-epoxyeicosatrienoic acid in cultured neurons from ASMko and WT mice. We found elevated concentrations of esterified pro-resolving lipid mediators and hydroxyeicosatrienoic acids typically destined for pro-resolving lipid mediator synthesis (e.g. lipoxins) in the cerebellum and neurons of ASMko mice compared to controls. Free d11-14(15)-epoxyeicosatrienoic acid esterification within neurons of ASMko mice was significantly elevated compared to WT. Our findings show evidence of increased inactivation of free pro-resolving lipid mediators through esterification in ASMko mice, suggesting impaired resolution as a new pathway underlying ASM deficiency pathogenesis.


Asunto(s)
Enfermedad de Niemann-Pick Tipo A , Enfermedades de Niemann-Pick , Animales , Ratones , Encéfalo/metabolismo , Esterificación , Inflamación/metabolismo , Ratones Noqueados , Neuronas/metabolismo , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/metabolismo , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedades de Niemann-Pick/metabolismo , Enfermedades de Niemann-Pick/patología , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielinas/metabolismo
2.
Nanomedicine ; 53: 102705, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37633404

RESUMEN

Acid sphingomyelinase deficiency (ASMD) is a severe lipid storage disorder caused by the diminished activity of the acid sphingomyelinase enzyme. ASMD is characterized by the accumulation of sphingomyelin in late endosomes and lysosomes leading to progressive neurological dysfunction and hepatosplenomegaly. Our objective was to investigate the utility of synthetic apolipoprotein A-I (ApoA-I) mimetics designed to act as lipid scavengers for the treatment of ASMD. We determined the lead peptide, 22A, could reduce sphingomyelin accumulation in ASMD patient skin fibroblasts in a dose dependent manner. Intraperitoneal administration of 22A formulated as a synthetic high-density lipoprotein (sHDL) nanodisc mobilized sphingomyelin from peripheral tissues into circulation and improved liver function in a mouse model of ASMD. Together, our data demonstrates that apolipoprotein mimetics could serve as a novel therapeutic strategy for modulating the pathology observed in ASMD.


Asunto(s)
Enfermedad de Niemann-Pick Tipo A , Animales , Ratones , Humanos , Enfermedad de Niemann-Pick Tipo A/tratamiento farmacológico , Enfermedad de Niemann-Pick Tipo A/patología , Esfingomielinas , Péptidos/uso terapéutico , Hígado/patología
3.
Cell Death Dis ; 14(4): 248, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-37024473

RESUMEN

Acid sphingomyelinase deficiency (ASMD) is a lysosomal storage disorder caused by mutations in the SMPD1 gene encoding for the acid sphingomyelinase (ASM). While intravenous infusion of recombinant ASM is an effective treatment for the peripheral disease, the neurological complications of ASMD remain unaddressed. It has been shown that aberrantly high level of total brain sphingomyelin (SM) is a key pathological event leading to neurodegeneration. Using mice lacking ASM (ASMko), which mimic the disease, we here demonstrate that among the SM species, SM16:0 shows the highest accumulation and toxicity in ASMko neurons. By targeting lysosomes, SM16:0 causes permeabilization and exocytosis of these organelles and induces oxidative stress and cell death. We also show that genetic silencing of Ceramide Synthase 5, which is involved in SM16:0 synthesis and overexpressed in the ASMko brain, prevents disease phenotypes in ASMko cultured neurons and mice. The levels of SM16:0 in plasma also show a strong correlation with those in brain that is higher than in liver, even at early stages of the disease. These results identify SM16:0 both as a novel therapeutic target and potential biomarker of brain pathology in ASMD.


Asunto(s)
Enfermedad de Niemann-Pick Tipo A , Ratones , Animales , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/metabolismo , Enfermedad de Niemann-Pick Tipo A/patología , Esfingomielinas/metabolismo , Ratones Noqueados , Esfingomielina Fosfodiesterasa/metabolismo , Encéfalo/metabolismo , Lisosomas/metabolismo
4.
Hereditas ; 160(1): 11, 2023 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-36907956

RESUMEN

BACKGROUND: Acid sphingomyelinase deficiency (ASMD) disorder, also known as Niemann-Pick disease (NPD) is a rare genetic disease caused by mutations in SMPD1 gene, which encodes sphingomyelin phosphodiesterase (ASM). Except for liver and spleen enlargement and lung disease, two subtypes (Type A and B) of NDP have different onset times, survival times, ASM activities, and neurological abnormalities. To comprehensively explore NPD's genotype-phenotype association and pathophysiological characteristics, we collected 144 NPD cases with strict quality control through literature mining. RESULTS: The difference in ASM activity can differentiate NPD type A from other subtypes, with the ratio of ASM activity to the reference values being lower in type A (threshold 0.045 (4.45%)). Severe variations, such as deletion and insertion, can cause complete loss of ASM function, leading to type A, whereas relatively mild missense mutations generally result in type B. Among reported mutations, the p.Arg3AlafsX76 mutation is highly prevalent in the Chinese population, and the p.R608del mutation is common in Mediterranean countries. The expression profiles of SMPD1 from GTEx and single-cell RNA sequencing data of multiple fetal tissues showed that high expressions of SMPD1 can be observed in the liver, spleen, and brain tissues of adults and hepatoblasts, hematopoietic stem cells, STC2_TLX1-positive cells, mesothelial cells of the spleen, vascular endothelial cells of the cerebellum and the cerebrum of fetuses, indicating that SMPD1 dysfunction is highly likely to have a significant effect on the function of those cell types during development and the clinicians need pay attention to these organs or tissues as well during diagnosis. In addition, we also predicted 21 new pathogenic mutations in the SMPD1 gene that potentially cause the NPD, signifying that more rare cases will be detected with those mutations in SMPD1. Finally, we also analysed the function of the NPD type A cells following the extracellular milieu. CONCLUSIONS: Our study is the first to elucidate the effects of SMPD1 mutation on cell types and at the tissue level, which provides new insights into the genotype-phenotype association and can help in the precise diagnosis of NPD.


Asunto(s)
Enfermedad de Niemann-Pick Tipo A , Enfermedades de Niemann-Pick , Esfingomielina Fosfodiesterasa , Humanos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Estudios de Asociación Genética , Mutación , Enfermedad de Niemann-Pick Tipo A/diagnóstico , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedades de Niemann-Pick/diagnóstico , Enfermedades de Niemann-Pick/genética , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo
5.
J Mol Neurosci ; 72(7): 1482-1499, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35727525

RESUMEN

Niemann-Pick type A disease (NPA) is a rare lysosomal storage disorder caused by mutations in the gene coding for the lysosomal enzyme acid sphingomyelinase (ASM). ASM deficiency leads to the consequent accumulation of its uncatabolized substrate, the sphingolipid sphingomyelin (SM), causing severe progressive brain disease. To study the effect of the aberrant lysosomal accumulation of SM on cell homeostasis, we loaded skin fibroblasts derived from a NPA patient with exogenous SM to mimic the levels of accumulation characteristic of the pathological neurons. In SM-loaded NPA fibroblasts, we found the blockage of the autophagy flux and the impairment of the mitochondrial compartment paralleled by the altered transcription of several genes, mainly belonging to the electron transport chain machinery and to the cholesterol biosynthesis pathway. In addition, SM loading induces the nuclear translocation of the transcription factor EB that promotes the lysosomal biogenesis and exocytosis. Interestingly, we obtained similar biochemical findings in the brain of the NPA mouse model lacking ASM (ASMKO mouse) at the neurodegenerative stage. Our work provides a new in vitro model to study NPA etiopathology and suggests the existence of a pathogenic lysosome-plasma membrane axis that with an impairment in the mitochondrial activity is responsible for the cell death.


Asunto(s)
Enfermedad de Niemann-Pick Tipo A , Enfermedades de Niemann-Pick , Animales , Apoptosis , Lisosomas/metabolismo , Ratones , Mitocondrias/metabolismo , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedades de Niemann-Pick/metabolismo , Enfermedades de Niemann-Pick/patología , Esfingomielinas/metabolismo , Esfingomielinas/farmacología
6.
Hum Mutat ; 42(10): 1336-1350, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34273913

RESUMEN

Pathogenic variations in SMPD1 lead to acid sphingomyelinase deficiency (ASMD), that is, Niemann-Pick disease (NPD) type A and B (NPA, NPB), which is a recessive lysosomal storage disease. The knowledge of variant spectrum in Indian patients is crucial for early and accurate NPD diagnosis and genetic counseling of families. In this study, we recruited 40 unrelated pediatric patients manifesting symptoms of ASMD and subnormal ASM enzyme activity. Variations in SMPD1 were studied using Sanger sequencing for all exons, followed by interpretation of variants based on American College of Medical Genetics and Genomics & Association for Molecular Pathology (ACMG/AMP) criteria. We identified 18 previously unreported variants and 21 known variants, including missense, nonsense, deletions, duplications, and splice site variations with disease-causing potential. Eight missense variants were functionally characterized using in silico molecular dynamic simulation and in vitro transient transfection in HEK293T cells, followed by ASM enzyme assay, immunoblot, and immunofluorescence studies. All the variants showed reduced ASM activity in transfected cells confirming their disease-causing potential. The study provides data for efficient prenatal diagnosis and genetic counseling of families with NPD type A and B.


Asunto(s)
Enfermedad de Niemann-Pick Tipo A , Enfermedades de Niemann-Pick , Esfingomielina Fosfodiesterasa/genética , Niño , Exones , Femenino , Células HEK293 , Humanos , Mutación , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedades de Niemann-Pick/diagnóstico , Enfermedades de Niemann-Pick/genética , Embarazo
7.
Am J Respir Cell Mol Biol ; 64(5): 629-640, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33662226

RESUMEN

Deficiency of ASM (acid sphingomyelinase) causes the lysosomal storage Niemann-Pick disease (NPD). Patients with NPD type B may develop progressive interstitial lung disease with frequent respiratory infections. Although several investigations using the ASM-deficient (ASMKO) mouse NPD model revealed inflammation and foamy macrophages, there is little insight into the pathogenesis of NPD-associated lung disease. Using ASMKO mice, we report that ASM deficiency is associated with a complex inflammatory phenotype characterized by marked accumulation of monocyte-derived CD11b+ macrophages and expansion of airspace/alveolar CD11c+ CD11b- macrophages, both with increased size, granularity, and foaminess. Both the alternative and classical pathways were activated, with decreased in situ phagocytosis of opsonized (Fc-coated) targets, preserved clearance of apoptotic cells (efferocytosis), secretion of Th2 cytokines, increased CD11c+/CD11b+ cells, and more than a twofold increase in lung and plasma proinflammatory cytokines. Macrophages, neutrophils, eosinophils, and noninflammatory lung cells of ASMKO lungs also exhibited marked accumulation of chitinase-like protein Ym1/2, which formed large eosinophilic polygonal Charcot-Leyden-like crystals. In addition to providing insight into novel features of lung inflammation that may be associated with NPD, our report provides a novel connection between ASM and the development of crystal-associated lung inflammation with alterations in macrophage biology.


Asunto(s)
Glicoproteínas/inmunología , Lisofosfolipasa/inmunología , Macrófagos Alveolares/inmunología , Macrófagos/inmunología , Enfermedad de Niemann-Pick Tipo A/inmunología , Enfermedad de Niemann-Pick Tipo B/inmunología , Neumonía/inmunología , Esfingomielina Fosfodiesterasa/inmunología , Animales , Antígenos CD11/genética , Antígenos CD11/inmunología , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Tamaño de la Célula , Quitinasas/genética , Quitinasas/inmunología , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Eosinófilos/patología , Femenino , Expresión Génica , Glicoproteínas/genética , Humanos , Lectinas/genética , Lectinas/inmunología , Pulmón/inmunología , Pulmón/patología , Lisofosfolipasa/genética , Macrófagos/patología , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Enfermedad de Niemann-Pick Tipo A/enzimología , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedad de Niemann-Pick Tipo B/enzimología , Enfermedad de Niemann-Pick Tipo B/genética , Enfermedad de Niemann-Pick Tipo B/patología , Fagocitosis , Neumonía/enzimología , Neumonía/genética , Neumonía/patología , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética , Balance Th1 - Th2/genética , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/inmunología
8.
Biochim Biophys Acta Mol Basis Dis ; 1866(10): 165875, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32522631

RESUMEN

Lysosomal storage disorders (LSDs) are diseases characterized by the accumulation of macromolecules in the late endocytic system and are caused by inherited defects in genes that encode mainly lysosomal enzymes or transmembrane lysosomal proteins. Niemann-Pick type C disease (NPCD), a LSD characterized by liver damage and progressive neurodegeneration that leads to early death, is caused by mutations in the genes encoding the NPC1 or NPC2 proteins. Both proteins are involved in the transport of cholesterol from the late endosomal compartment to the rest of the cell. Loss of function of these proteins causes primary cholesterol accumulation, and secondary accumulation of other lipids, such as sphingolipids, in lysosomes. Despite years of studying the genetic and molecular bases of NPCD and related-lysosomal disorders, the pathogenic mechanisms involved in these diseases are not fully understood. In this review we will summarize the pathogenic mechanisms described for NPCD and we will discuss their relevance for other LSDs with neurological components such as Niemann- Pick type A and Gaucher diseases. We will particularly focus on the activation of signaling pathways that may be common to these three pathologies with emphasis on how the intra-lysosomal accumulation of lipids leads to pathology, specifically to neurological impairments. We will show that although the primary lipid storage defect is different in these three LSDs, there is a similar secondary accumulation of metabolites and activation of signaling pathways that can lead to common pathogenic mechanisms. This analysis might help to delineate common pathological mechanisms and therapeutic targets for lysosomal storage diseases.


Asunto(s)
Enfermedad de Gaucher/metabolismo , Metabolismo de los Lípidos/genética , Lisosomas/patología , Enfermedad de Niemann-Pick Tipo A/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/patología , Colesterol/metabolismo , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/patología , Glucosilceramidasa/genética , Glucosilceramidasa/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/metabolismo , Mutación , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/patología , Transducción de Señal/genética , Esfingolípidos/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
9.
Ann Hepatol ; 18(4): 613-619, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31122880

RESUMEN

INTRODUCTION AND OBJECTIVES: Niemann-Pick disease type A (NPD-A) and B (NPD-B) are lysosomal storage diseases with a birth prevalence of 0.4-0.6/100,000. They are caused by a deficiency in acid sphingomyelinase, an enzyme encoded by SMPD1. We analyzed the phenotype and genotype of four unrelated Mexican patients, one with NPD-A and three with NPD-B. PATIENTS AND METHODS: Four female patients between 1 and 7 years of age were diagnosed with NPD-A or NPD-B by hepatosplenomegaly, among other clinical characteristics, and by determining the level of acid sphingomyelinase enzymatic activity and sequencing of the SMPD1 gene. Additionally, a 775bp amplicon of SMPD1 (from 11:6393835_6394609, including exons 5 and 6) was analyzed by capillary sequencing in a control group of 50 unrelated healthy Mexican Mestizos. RESULTS: An infrequent variant (c.1343A>G p.Tyr448Cys) was observed in two patients. One is the first NPD-A homozygous patient reported with this variant and the other a compound heterozygous NPD-B patient with the c.1829_1831delGCC p.Arg610del variant. Another compound heterozygous patient had the c.1547A>G p.His516Arg variant (not previously described in affected individuals) along with the c.1805G>A p.Arg602His variant. A new c.1263+8C>T pathogenic variant was encountered in a homozygous state in a NPD-B patient. Among the healthy control individuals there was a heterozygous carrier for the c.1550A>T (rs142787001) pathogenic variant, but none with the known pathogenic variants in the 11:6393835_6394609 region of SMPD1. CONCLUSIONS: The present study provides further NPD-A or B phenotype-genotype correlations. We detected a heterozygous carrier with a pathogenic variant in 1/50 healthy Mexican mestizos.


Asunto(s)
Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo B/genética , Esfingomielina Fosfodiesterasa/genética , Adolescente , Adulto , Niño , Preescolar , Epistaxis/fisiopatología , Femenino , Tamización de Portadores Genéticos , Genotipo , Trastornos del Crecimiento/fisiopatología , Voluntarios Sanos , Hepatomegalia/fisiopatología , Heterocigoto , Humanos , Lactante , Hígado/patología , Hígado/ultraestructura , México , Enfermedad de Niemann-Pick Tipo A/metabolismo , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedad de Niemann-Pick Tipo A/fisiopatología , Enfermedad de Niemann-Pick Tipo B/metabolismo , Enfermedad de Niemann-Pick Tipo B/patología , Enfermedad de Niemann-Pick Tipo B/fisiopatología , Fenotipo , Esfingomielina Fosfodiesterasa/metabolismo , Esplenomegalia/fisiopatología , Adulto Joven
10.
Stem Cell Res ; 38: 101461, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31132580

RESUMEN

Niemann-Pick disease type A (NPA) is a rare autosomal recessive lysosomal storage disease caused by mutations in the SMPD1 gene, which encodes for the protein acid sphingomyelinase. A human induced pluripotent stem cell (iPSC) line was generated from dermal fibroblasts of a 21-fetal-week-old female patient with NPA that has a heterozygous mutation of a p.L302P variant (c.905 T > C) using non-integrating Sendai virus technique. This iPSC line offers a useful resource to study the disease pathophysiology and as a cell-based model for drug development to treat NPA.


Asunto(s)
Técnicas de Reprogramación Celular , Células Madre Pluripotentes Inducidas , Mutación Missense , Enfermedad de Niemann-Pick Tipo A , Esfingomielina Fosfodiesterasa , Sustitución de Aminoácidos , Línea Celular , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/patología , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo
11.
EMBO J ; 38(2)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30530526

RESUMEN

Neuropathic lysosomal storage disorders (LSDs) present with activated pro-inflammatory microglia. However, anti-inflammatory treatment failed to improve disease pathology. We characterise the mechanisms underlying microglia activation in Niemann-Pick disease type A (NPA). We establish that an NPA patient and the acid sphingomyelinase knockout (ASMko) mouse model show amoeboid microglia in neurodegeneration-prone areas. In vivo microglia ablation worsens disease progression in ASMko mice. We demonstrate the coexistence of different microglia phenotypes in ASMko brains that produce cytokines or counteract neuronal death by clearing myelin debris. Overloading microglial lysosomes through myelin debris accumulation and sphingomyelin build-up induces lysosomal damage and cathepsin B extracellular release by lysosomal exocytosis. Inhibition of cathepsin B prevents neuronal death and behavioural anomalies in ASMko mice. Similar microglia phenotypes occur in a Niemann-Pick disease type C mouse model and patient. Our results show a protective function for microglia in LSDs and how this is corrupted by lipid lysosomal overload. Data indicate cathepsin B as a key molecule mediating neurodegeneration, opening research pathways for therapeutic targeting of LSDs and other demyelinating diseases.


Asunto(s)
Catepsina B/metabolismo , Microglía/patología , Enfermedad de Niemann-Pick Tipo A/patología , Esfingomielina Fosfodiesterasa/genética , Animales , Línea Celular , Preescolar , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Recién Nacido , Lisosomas/metabolismo , Lisosomas/patología , Ratones , Ratones Noqueados , Microglía/metabolismo , Enfermedad de Niemann-Pick Tipo A/genética , Fenotipo , Esfingomielinas/metabolismo
12.
Clin Biochem ; 61: 40-44, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30205089

RESUMEN

OBJECTIVE: Plasma chitotriosidase is a documented biomarker for certain lysosomal storage disorders. However, its clinical utility for prenatal samples is not elucidated yet. METHODS: We have established Reference intervals for amniotic fluid chitotriosidase using control amniotic fluids (n = 47) and compared the activity with amniotic fluids affected by lysosomal storage disorders (n = 25). RESULTS: The reference interval established was 0-6.76 nmol/h/ml. The amniotic fluids affected with LSDs exhibited elevation of chitotriosidase. The area under the curve (AUC) of receiver operating characteristic curve for affected vs. healthy was 0.987 indicating 98.6% accuracy of chitotriosidase in identifying pregnancies affected with LSDs. Among the different LSDs, Gaucher (202.00 ±â€¯35.27 nmol/h/ml) and Niemann-pick A/B (60.33 ±â€¯21.59 nmol/h/ml) showed very high levels of chitotriosidase. CONCLUSION: Amniotic fluid chitotriosidase has the potential to serve as a diagnostic marker for lysosomal storage disorders, more specifically for Gaucher and Niemann-Pick A/B.


Asunto(s)
Amniocentesis , Líquido Amniótico/enzimología , Hexosaminidasas/metabolismo , Enfermedades por Almacenamiento Lisosomal/diagnóstico , Regulación hacia Arriba , Adulto , Área Bajo la Curva , Biomarcadores/metabolismo , Células Cultivadas , Estudios de Cohortes , Exones , Femenino , Enfermedad de Gaucher/diagnóstico , Enfermedad de Gaucher/enzimología , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/patología , Duplicación de Gen , Hexosaminidasas/química , Hexosaminidasas/genética , Humanos , India , Enfermedades por Almacenamiento Lisosomal/enzimología , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/patología , Tasa de Mutación , Enfermedad de Niemann-Pick Tipo A/diagnóstico , Enfermedad de Niemann-Pick Tipo A/enzimología , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedad de Niemann-Pick Tipo B/diagnóstico , Enfermedad de Niemann-Pick Tipo B/enzimología , Enfermedad de Niemann-Pick Tipo B/genética , Enfermedad de Niemann-Pick Tipo B/patología , Embarazo , Segundo Trimestre del Embarazo , Curva ROC , Valores de Referencia , Adulto Joven
13.
Mol Psychiatry ; 22(5): 711-723, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27620840

RESUMEN

Niemann-Pick disease type A (NPA) is a rare lysosomal storage disorder characterized by severe neurological alterations that leads to death in childhood. Loss-of-function mutations in the acid sphingomyelinase (ASM) gene cause NPA, and result in the accumulation of sphingomyelin (SM) in lysosomes and plasma membrane of neurons. Using ASM knockout (ASMko) mice as a NPA disease model, we investigated how high SM levels contribute to neural pathology in NPA. We found high levels of oxidative stress both in neurons from these mice and a NPA patient. Impaired activity of the plasma membrane calcium ATPase (PMCA) increases intracellular calcium. SM induces PMCA decreased activity, which causes oxidative stress. Incubating ASMko-cultured neurons in the histone deacetylase inhibitor, SAHA, restores PMCA activity and calcium homeostasis and, consequently, reduces the increased levels of oxidative stress. No recovery occurs when PMCA activity is pharmacologically impaired or genetically inhibited in vitro. Oral administration of SAHA prevents oxidative stress and neurodegeneration, and improves behavioral performance in ASMko mice. These results demonstrate a critical role for plasma membrane SM in neuronal calcium regulation. Thus, we identify changes in PMCA-triggered calcium homeostasis as an upstream mediator for NPA pathology. These findings can stimulate new approaches for pharmacological remediation in a disease with no current clinical treatments.


Asunto(s)
Enfermedad de Niemann-Pick Tipo A/metabolismo , Enfermedad de Niemann-Pick Tipo A/patología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/antagonistas & inhibidores , Esfingomielinas/metabolismo , Animales , Encéfalo/metabolismo , Estudios de Casos y Controles , Membrana Celular/enzimología , Membrana Celular/metabolismo , Preescolar , Modelos Animales de Enfermedad , Humanos , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/enzimología , Neuronas/metabolismo , Enfermedad de Niemann-Pick Tipo A/enzimología , Estrés Oxidativo/fisiología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo
15.
Stem Cells Transl Med ; 5(12): 1644-1655, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27484861

RESUMEN

: Niemann-Pick disease type A (NPA) is a lysosomal storage disease caused by mutations in the SMPD1 gene that encodes acid sphingomyelinase (ASM). Deficiency in ASM function results in lysosomal accumulation of sphingomyelin and neurodegeneration. Currently, there is no effective treatment for NPA. To accelerate drug discovery for treatment of NPA, we generated induced pluripotent stem cells from two patient dermal fibroblast lines and differentiated them into neural stem cells. The NPA neural stem cells exhibit a disease phenotype of lysosomal sphingomyelin accumulation and enlarged lysosomes. By using this disease model, we also evaluated three compounds that reportedly reduced lysosomal lipid accumulation in Niemann-Pick disease type C as well as enzyme replacement therapy with ASM. We found that α-tocopherol, δ-tocopherol, hydroxypropyl-ß-cyclodextrin, and ASM reduced sphingomyelin accumulation and enlarged lysosomes in NPA neural stem cells. Therefore, the NPA neural stem cells possess the characteristic NPA disease phenotype that can be ameliorated by tocopherols, cyclodextrin, and ASM. Our results demonstrate the efficacies of cyclodextrin and tocopherols in the NPA cell-based model. Our data also indicate that the NPA neural stem cells can be used as a new cell-based disease model for further study of disease pathophysiology and for high-throughput screening to identify new lead compounds for drug development. SIGNIFICANCE: Currently, there is no effective treatment for Niemann-Pick disease type A (NPA). To accelerate drug discovery for treatment of NPA, NPA-induced pluripotent stem cells were generated from patient dermal fibroblasts and differentiated into neural stem cells. By using the differentiated NPA neuronal cells as a cell-based disease model system, α-tocopherol, δ-tocopherol, and hydroxypropyl-ß-cyclodextrin significantly reduced sphingomyelin accumulation in these NPA neuronal cells. Therefore, this cell-based NPA model can be used for further study of disease pathophysiology and for high-throughput screening of compound libraries to identify lead compounds for drug development.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedad de Niemann-Pick Tipo A/terapia , 2-Hidroxipropil-beta-Ciclodextrina , Diferenciación Celular/efectos de los fármacos , Línea Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibroblastos/ultraestructura , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/ultraestructura , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/ultraestructura , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Células-Madre Neurales/ultraestructura , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielinas/metabolismo , Tocoferoles/farmacología , alfa-Tocoferol/farmacología , beta-Ciclodextrinas/farmacología
16.
Autophagy ; 12(5): 833-49, 2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-27070082

RESUMEN

Sphingomyelin is an essential cellular lipid that traffics between plasma membrane and intracellular organelles until directed to lysosomes for SMPD1 (sphingomyelin phosphodiesterase 1)-mediated degradation. Inactivating mutations in the SMPD1 gene result in Niemann-Pick diseases type A and B characterized by sphingomyelin accumulation and severely disturbed tissue homeostasis. Here, we report that sphingomyelin overload disturbs the maturation and closure of autophagic membranes. Niemann-Pick type A patient fibroblasts and SMPD1-depleted cancer cells accumulate elongated and unclosed autophagic membranes as well as abnormally swollen autophagosomes in the absence of normal autophagosomes and autolysosomes. The immature autophagic membranes are rich in WIPI2, ATG16L1 and MAP1LC3B but display reduced association with ATG9A. Contrary to its normal trafficking between plasma membrane, intracellular organelles and autophagic membranes, ATG9A concentrates in transferrin receptor-positive juxtanuclear recycling endosomes in SMPD1-deficient cells. Supporting a causative role for ATG9A mistrafficking in the autophagy defect observed in SMPD1-deficient cells, ectopic ATG9A effectively reverts this phenotype. Exogenous C12-sphingomyelin induces a similar juxtanuclear accumulation of ATG9A and subsequent defect in the maturation of autophagic membranes in healthy cells while the main sphingomyelin metabolite, ceramide, fails to revert the autophagy defective phenotype in SMPD1-deficient cells. Juxtanuclear accumulation of ATG9A and defective autophagy are also evident in tissues of smpd1-deficient mice with a subsequent inability to cope with kidney ischemia-reperfusion stress. These data reveal sphingomyelin as an important regulator of ATG9A trafficking and maturation of early autophagic membranes.


Asunto(s)
Autofagosomas/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia/fisiología , Proteínas de la Membrana/metabolismo , Esfingomielinas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Autofagosomas/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Membrana Celular/metabolismo , Endosomas/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Células MCF-7 , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/metabolismo , Enfermedad de Niemann-Pick Tipo A/patología , Transporte de Proteínas , ARN Interferente Pequeño/genética , Receptores de Transferrina/metabolismo , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética
17.
Hum Mutat ; 37(2): 139-47, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26499107

RESUMEN

Niemann-Pick Types A and B (NPA/B) diseases are autosomal recessive lysosomal storage disorders caused by the deficient activity of acid sphingomyelinase (ASM) because of the mutations in the SMPD1 gene. Here, we provide a comprehensive updated review of already reported and newly identified SMPD1 variants. Among them, 185 have been found in NPA/B patients. Disease-causing variants are equally distributed along the SMPD1 gene; most of them are missense (65.4%) or frameshift (19%) mutations. The most frequently reported mutation worldwide is the p.R610del, clearly associated with an attenuated NP disease type B phenotype. The available information about the impact of 52 SMPD1 variants on ASM mRNA and/or enzymatic activity has been collected and whenever possible, phenotype/genotype correlations were established. In addition, we created a locus-specific database easily accessible at http://www.inpdr.org/genes that catalogs the 417 SMPD1 variants reported to date and provides data on their in silico predicted effects on ASM protein function or mRNA splicing. The information reviewed in this article, providing new insights into the genotype/phenotype correlation, is extremely valuable to facilitate diagnosis and genetic counseling of families affected by NPA/B.


Asunto(s)
Bases de Datos Genéticas , Mutación , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo B/genética , ARN Mensajero/genética , Esfingomielina Fosfodiesterasa/genética , Exones , Expresión Génica , Genes Recesivos , Estudios de Asociación Genética , Genotipo , Humanos , Intrones , Enfermedad de Niemann-Pick Tipo A/diagnóstico , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedad de Niemann-Pick Tipo B/diagnóstico , Enfermedad de Niemann-Pick Tipo B/patología , Sistemas de Lectura Abierta , Fenotipo , Empalme del ARN
18.
Neurobiol Dis ; 73: 319-26, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25251606

RESUMEN

Niemann-Pick disease type A (NPDA) is a fatal disease due to mutations in the acid sphingomyelinase (ASM) gene, which triggers the abnormal accumulation of sphingomyelin (SM) in lysosomes and the plasma membrane of mutant cells. Although the disease affects multiple organs, the impact on the brain is the most invalidating feature. The mechanisms responsible for the cognitive deficit characteristic of this condition are only partially understood. Using mice lacking the ASM gene (ASMko), a model system in NPDA research, we report here that high sphingomyelin levels in mutant neurons lead to low synaptic levels of phosphoinositide PI(4,5)P2 and reduced activity of its hydrolyzing phosphatase PLCγ, which are key players in synaptic plasticity events. In addition, mutant neurons have reduced levels of membrane-bound MARCKS, a protein required for PI(4,5)P2 membrane clustering and hydrolysis. Intracerebroventricular infusion of a peptide that mimics the effector domain of MARCKS increases the content of PI(4,5)P2 in the synaptic membrane and ameliorates behavioral abnormalities in ASMko mice.


Asunto(s)
Encéfalo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/uso terapéutico , Proteínas de la Membrana/uso terapéutico , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/etiología , Enfermedad de Niemann-Pick Tipo A/complicaciones , Enfermedad de Niemann-Pick Tipo A/tratamiento farmacológico , Animales , Reacción de Prevención/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Inyecciones Intraventriculares , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Fuerza Muscular/efectos de los fármacos , Fuerza Muscular/genética , Mutación/genética , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Enfermedad de Niemann-Pick Tipo A/metabolismo , Enfermedad de Niemann-Pick Tipo A/patología , Fosfolipasa C gamma/metabolismo , Esfingomielina Fosfodiesterasa/genética , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo
19.
Cell Death Differ ; 21(6): 864-75, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24488099

RESUMEN

Niemann Pick disease type A (NPA), which is caused by loss of function mutations in the acid sphingomyelinase (ASM) gene, is a lysosomal storage disorder leading to neurodegeneration. Yet, lysosomal dysfunction and its consequences in the disease are poorly characterized. Here we show that undegraded molecules build up in neurons of acid sphingomyelinase knockout mice and in fibroblasts from NPA patients in which autophagolysosomes accumulate. The latter is not due to alterations in autophagy initiation or autophagosome-lysosome fusion but because of inefficient autophago-lysosomal clearance. This, in turn, can be explained by lysosomal membrane permeabilization leading to cytosolic release of Cathepsin B. High sphingomyelin (SM) levels account for these effects as they can be induced in control cells on addition of the lipid and reverted on SM-lowering strategies in ASM-deficient cells. These results unveil a relevant role for SM in autophagy modulation and characterize autophagy anomalies in NPA, opening new perspectives for therapeutic interventions.


Asunto(s)
Autofagia/genética , Lisosomas/metabolismo , Enfermedad de Niemann-Pick Tipo A/genética , Esfingomielinas/metabolismo , Animales , Catepsina B/metabolismo , Humanos , Lisosomas/patología , Ratones , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedad de Niemann-Pick Tipo A/terapia , Esfingomielina Fosfodiesterasa/metabolismo
20.
EMBO Mol Med ; 6(3): 398-413, 2014 03.
Artículo en Inglés | MEDLINE | ID: mdl-24448491

RESUMEN

Understanding the role of lipids in synapses and the aberrant molecular mechanisms causing the cognitive deficits that characterize most lipidosis is necessary to develop therapies for these diseases. Here we describe sphingomyelin (SM) as a key modulator of the dendritic spine actin cytoskeleton. We show that increased SM levels in neurons of acid sphingomyelinase knock out mice (ASMko), which mimic Niemann Pick disease type A (NPA), result in reduced spine number and size and low levels of filamentous actin. Mechanistically, SM accumulation decreases the levels of metabotropic glutamate receptors type I (mGluR1/5) at the synaptic membrane impairing membrane attachment and activity of RhoA and its effectors ROCK and ProfilinIIa. Pharmacological enhancement of the neutral sphingomyelinase rescues the aberrant molecular and morphological phenotypes in vitro and in vivo and improves motor and memory deficits in ASMko mice. Altogether, these data demonstrate the influence of SM and its catabolic enzymes in dendritic spine physiology and contribute to our understanding of the cognitive deficits of NPA patients, opening new perspectives for therapeutic interventions.


Asunto(s)
Espinas Dendríticas/efectos de los fármacos , Enfermedad de Niemann-Pick Tipo A/tratamiento farmacológico , Enfermedad de Niemann-Pick Tipo A/patología , Citoesqueleto de Actina/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Espinas Dendríticas/metabolismo , Dexametasona/farmacología , Modelos Animales de Enfermedad , Femenino , Memoria a Corto Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedad de Niemann-Pick Tipo A/metabolismo , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielinas/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...