Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Appl Toxicol ; 41(10): 1584-1597, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33559204

RESUMEN

Silicosis is a serious occupational disease with the highest incidence in China. However, its pathogenesis has not been fully elucidated. Studies have shown that the sphingomyelin signaling pathway may play an important role in different fibrotic diseases but its role in silicosis-mediated fibrosis is still unclear. In this study, the supernatant of human peripheral blood mononuclear cell line (THP-1)-derived macrophages exposed to silica (SiO2 ) was used to stimulate the transformation of human embryonic lung fibroblast cell line (HFL-1) into myofibroblasts, and the intervention effect of recombinant human acid ceramidase (rAC) was observed. The results showed that SiO2 stimulated the production of reactive oxygen species and malondialdehyde in the supernatant of THP-1-derived macrophages and increased the secretion of TGF-ß1, TNF-α, and IL-8. In addition, we found that the expression levels of α-SMA, FN, Col I, and Col III in HFL-1 cells increased. Meanwhile, the activities of ASMase and ACase and the expression levels of Cer, Sph, and S1P were increased. Intervention by rAC can suppress these changes to different degrees. In conclusion, the present study shows that SiO2 dust poisoning may stimulate HFL-1 cell differentiation into myofibroblasts by inducing oxidative stress in THP-1-derived macrophages, thereby promoting the secretion of a variety of inflammatory factors and activating the sphingolipid signaling pathway in HFL-1 cells. Exogenous rAC can effectively interfere with the stimulation of HFL-1 cells by silica in vitro.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Dióxido de Silicio/metabolismo , Dióxido de Silicio/toxicidad , Silicosis/fisiopatología , Esfingomielinas/metabolismo , Adulto , China/epidemiología , Femenino , Humanos , Incidencia , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Redes y Vías Metabólicas , Persona de Mediana Edad , Enfermedades Profesionales/epidemiología , Enfermedades Profesionales/fisiopatología , Silicosis/epidemiología
2.
Nat Immunol ; 9(1): 42-53, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18037890

RESUMEN

Although much is known about the migration of T cells from blood to lymph nodes, less is known about the mechanisms regulating the migration of T cells from tissues into lymph nodes through afferent lymphatics. Here we investigated T cell egress from nonlymphoid tissues into afferent lymph in vivo and developed an experimental model to recapitulate this process in vitro. Agonism of sphingosine 1-phosphate receptor 1 inhibited the entry of tissue T cells into afferent lymphatics in homeostatic and inflammatory conditions and caused the arrest, mediated at least partially by interactions of the integrin LFA-1 with its ligand ICAM-1 and of the integrin VLA-4 with its ligand VCAM-1, of polarized T cells at the basal surface of lymphatic but not blood vessel endothelium. Thus, the increased sphingosine 1-phosphate present in inflamed peripheral tissues may induce T cell retention and suppress T cell egress.


Asunto(s)
Vasos Linfáticos/inmunología , Lisofosfolípidos/metabolismo , Modelos Inmunológicos , Receptores de Lisoesfingolípidos/inmunología , Esfingosina/análogos & derivados , Linfocitos T/inmunología , Animales , Movimiento Celular , Endotelio Linfático/inmunología , Clorhidrato de Fingolimod , Homeostasis , Inflamación/inmunología , Integrina alfa4beta1/inmunología , Molécula 1 de Adhesión Intercelular/inmunología , Ganglios Linfáticos/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Ratones , Ratones Endogámicos C57BL , Glicoles de Propileno/farmacología , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Transducción de Señal , Esfingosina/metabolismo , Esfingosina/farmacología , Molécula 1 de Adhesión Celular Vascular/inmunología
3.
Biochim Biophys Acta Mol Basis Dis ; 1863(2): 386-394, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27915031

RESUMEN

Acid Ceramidase Deficiency (Farber disease, FD) is an ultra-rare Lysosomal Storage Disorder that is poorly understood and often misdiagnosed as Juvenile Idiopathic Arthritis (JIA). Hallmarks of FD are accumulation of ceramides, widespread macrophage infiltration, splenomegaly, and lymphocytosis. The cytokines involved in this abnormal hematopoietic state are unknown. There are dozens of ceramide species and derivatives, but the specific ones that accumulate in FD have not been investigated. We used a multiplex assay to analyze cytokines and mass spectrometry to analyze ceramides in plasma from patients and mice with FD, controls, Farber patients treated by hematopoietic stem cell transplantation (HSCT), JIA patients, and patients with Gaucher disease. KC, MIP-1α, and MCP-1 were sequentially upregulated in plasma from FD mice. MCP-1, IL-10, IL-6, IL-12, and VEGF levels were elevated in plasma from Farber patients but not in control or JIA patients. C16-Ceramide (C16-Cer) and dhC16-Cer were upregulated in plasma from FD mice. a-OH-C18-Cer, dhC12-Cer, dhC24:1-Cer, and C22:1-Cer-1P accumulated in plasma from patients with FD. Most cytokines and only a-OH-C18-Cer returned to baseline levels in HSCT-treated Farber patients. Sphingosines were not altered. Chitotriosidase activity was also relatively low. A unique cytokine and ceramide profile was seen in the plasma of Farber patients that was not observed in plasma from HSCT-treated Farber patients, JIA patients, or Gaucher patients. The cytokine profile can potentially be used to prevent misdiagnosis of Farber as JIA and to monitor the response to treatment. Further understanding of why these signaling molecules and lipids are elevated can lead to better understanding of the etiology and pathophysiology of FD and inform development of future treatments.


Asunto(s)
Ceramidas/sangre , Citocinas/sangre , Lipogranulomatosis de Farber/sangre , Animales , Artritis Juvenil/sangre , Trasplante de Médula Ósea , Lipogranulomatosis de Farber/terapia , Femenino , Hexosaminidasas/sangre , Humanos , Masculino , Ratones
4.
J Lipid Res ; 57(9): 1696-711, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27474222

RESUMEN

We aimed to characterize the lipidomic, metabolomic, and transcriptomic profiles in preterm piglets administered enteral (ENT) formula or three parenteral lipid emulsions [parenteral nutrition (PN)], Intralipid (IL), Omegaven (OV), or SMOFlipid (SL), for 14 days. Piglets in all parenteral lipid groups showed differential organ growth versus ENT piglets; whole body growth rate was lowest in IL piglets, yet there were no differences in either energy expenditure or (13)C-palmitate oxidation. Plasma homeostatic model assessment of insulin resistance demonstrated insulin resistance in IL, but not OV or SL, compared with ENT. The fatty acid and acyl-CoA content of the liver, muscle, brain, and plasma fatty acids reflected the composition of the dietary lipids administered. Free carnitine and acylcarnitine (ACT) levels were markedly reduced in the PN groups compared with ENT piglets. Genes associated with oxidative stress and inflammation were increased, whereas those associated with alternative pathways of fatty acid oxidation were decreased in all PN groups. Our results show that new generation lipid emulsions directly enrich tissue fatty acids, especially in the brain, and lead to improved growth and insulin sensitivity compared with a soybean lipid emulsion. In all total PN groups, carnitine levels are limiting to the formation of ACTs and gene expression reflects the stress of excess lipid on liver function.


Asunto(s)
Aceites de Pescado/administración & dosificación , Metabolismo de los Lípidos/genética , Fosfolípidos/administración & dosificación , Aceite de Soja/administración & dosificación , Triglicéridos/metabolismo , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Animales Recién Nacidos/metabolismo , Grasas Insaturadas en la Dieta/administración & dosificación , Emulsiones/administración & dosificación , Ácidos Grasos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ácido Palmítico/administración & dosificación , Ácido Palmítico/metabolismo , Nutrición Parenteral , Porcinos/crecimiento & desarrollo , Porcinos/metabolismo
5.
Biol Chem ; 396(6-7): 759-65, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25938220

RESUMEN

Acid ceramidase (N-acylsphingosine deacylase, EC 3.5.1.23; AC) is the lipid hydrolase responsible for the degradation of ceramide into sphingosine and free fatty acids within lysosomes. The enzymatic activity was first identified over four decades ago and is deficient in two rare inherited disorders, Farber lipogranulomatosis (Farber disease) and spinal muscular atrophy with myoclonic epilepsy (SMA-PME). Importantly, AC not only hydrolyzes ceramide into sphingosine within acidic compartments, but also can synthesize ceramide from sphingosine at neutral pH, suggesting that the enzyme may have diverse functions depending on its subcellular location and the local pH. Within cells, AC exists in a complex with other lipid hydrolases and requires a polypeptide cofactor (saposin D) for full hydrolytic activity. Recent studies also have shown that AC is overexpressed in several human cancers, and that inhibition of this enzyme may be a useful cancer drug target. Aberrant AC activity has also been described in several other common diseases. The cDNA and gene (ASAH1) encoding AC have been isolated, several mouse models of AC deficiency have been constructed, and the recombinant enzyme is currently being manufactured for the treatment of Farber disease and SMA-PME. Current information concerning the biology of this enzyme and its role in human disease is reviewed within.


Asunto(s)
Ceramidasa Ácida/metabolismo , Medicina Molecular , Animales , Ceramidas/metabolismo , Terapia de Reemplazo Enzimático , Humanos , Lisosomas/metabolismo
6.
Proc Natl Acad Sci U S A ; 107(1): 222-7, 2010 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-20018674

RESUMEN

Enzyme replacement therapy is currently available for three of the mucopolysaccharidoses (MPSs) but has limited effects on the skeletal lesions. We investigated the involvement of the Toll-like receptor 4 (TLR4) signaling pathway in the pathogenesis of MPS bone and joint disease, and the use of the anti-TNF-alpha drug, Remicade (Centocor, Inc.), for treatment. TLR4 KO (TLR4(lps-/-)) mice were interbred with MPS VII mice to produce double-KO (DKO) animals. The DKO mice had longer and thinner faces and longer femora as revealed by micro-computed tomography analysis compared with MPS VII mice. Histological analyses also revealed more organized and thinner growth plates. The serum levels of TNF-alpha were normalized in the DKO animals, and the levels of phosphorylated STAT1 and STAT3 in articular chondrocytes were corrected. These findings led us to evaluate the effects of Remicade in MPS VI rats. When initiated at 1 month of age, i.v. treatment prevented the elevation of TNF-alpha, receptor activator of NF-kappaB, and other inflammatory molecules not only in the blood but in articular chondrocytes and fibroblast-like synoviocytes (FLSs). Treatment of 6-month-old animals also reduced the levels of these molecules to normal. The number of apoptotic articular chondrocytes in MPS VI rats was similarly reduced, with less infiltration of synovial tissue into the underlying bone. These studies revealed the important role of TLR4 signaling in MPS bone and joint disease and suggest that targeting TNF-alpha may have positive therapeutic effects.


Asunto(s)
Mucopolisacaridosis/tratamiento farmacológico , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Huesos/anatomía & histología , Huesos/diagnóstico por imagen , Huesos/metabolismo , Huesos/patología , Humanos , Infliximab , Masculino , Ratones , Ratones Noqueados , Mucopolisacaridosis/inmunología , Mucopolisacaridosis/patología , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología , Microtomografía por Rayos X
7.
Biomolecules ; 13(11)2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-38002305

RESUMEN

Acid ceramidase (AC) is a lysosomal enzyme required to hydrolyze ceramide to sphingosine by the removal of the fatty acid moiety. An inherited deficiency in this activity results in two disorders, Farber Lipogranulomatosis and spinal muscular atrophy with myoclonic epilepsy, leading to the accumulation of ceramides and other sphingolipids in various cells and tissues. In addition to ceramide hydrolysis, several other activities have been attributed to AC, including a reverse reaction that synthesizes ceramide from free fatty acids and sphingosine, and a deacylase activity that removes fatty acids from complex lipids such as sphingomyelin and glycosphingolipids. A close association of AC with another important enzyme of sphingolipid metabolism, acid sphingomyelinase (ASM), has also been observed. Herein, we used a highly purified recombinant human AC (rhAC) and novel UPLC-based assay methods to investigate the recently described deacylase activity of rhAC against three sphingolipid substrates, sphingomyelin, galactosyl- and glucosylceramide. No deacylase activities were detected using this method, although we did unexpectedly identify a significant ASM activity using natural (C-18) and artificial (Bodipy-C12) sphingomyelin substrates as well as the ASM-specific fluorogenic substrate, hexadecanoylamino-4-methylumbelliferyl phosphorylcholine (HMU-PC). We showed that this ASM activity was not due to contaminating, hamster-derived ASM in the rhAC preparation, and that the treatment of ASM-knockout mice with rhAC significantly reduced sphingomyelin storage in the liver. However, unlike the treatment with rhASM, this did not lead to elevated ceramide or sphingosine levels.


Asunto(s)
Ceramidasa Ácida , Esfingomielinas , Animales , Ratones , Cricetinae , Humanos , Ceramidasa Ácida/genética , Ceramidasa Ácida/metabolismo , Esfingomielinas/metabolismo , Esfingosina/metabolismo , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Ceramidas/metabolismo , Esfingolípidos/metabolismo , Ácidos Grasos
8.
J Biol Chem ; 286(41): 35624-35633, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21846728

RESUMEN

Autoproteolytic cleavage of the inactive acid ceramidase (AC) precursor into the active heterodimer exposes a free cysteine residue, leading us to study whether AC could be regulated by one or more members of the cystatin family. Co-expression of the full-length AC and cystatin SA (cysSA) cDNAs led to significant reduction of AC activity in the transfected cells. Expression of cysSA also inhibited endogenous AC activity in cells and increased ceramide. Conversely, cysSA siRNA expression led to elevated AC activity and reduction in ceramide. The effects of cysSA siRNA expression could be reversed by the addition of recombinant cysSA into the culture media. These results were consistent with detection of a physical interaction between AC and cysSA, assessed by co-immunoprecipitation and nickel-nitrilotriacetic acid affinity chromatography, and further supported by co-localization of the endogenous proteins using confocal microscopy. In vitro kinetic analysis of purified, recombinant AC and cysSA confirmed the transfection results and suggested a non-competitive type of inhibition with a K(i) in the low micromolar range. Processing of the AC precursor into the active form was not affected by cysSA expression, suggesting that it likely inhibits AC by allosteric interference. Computer modeling and expression studies identified several potential inhibitory domains in cysSA, including a small "AC-like" domain (identical to the AC cleavage site, TICT). Small peptides, synthesized with combinations of this and a "cystatin-like" domain (QXVXG), exhibited significant AC inhibition as well. Such peptide-based AC inhibitors could potentially be used to regulate AC activity in cancer cells that are known to overexpress this enzyme alone and in combination with conventional anti-cancer drugs.


Asunto(s)
Ceramidasa Ácida/antagonistas & inhibidores , Cistatina A/farmacología , Inhibidores Enzimáticos/farmacología , Ceramidasa Ácida/genética , Ceramidasa Ácida/metabolismo , Antineoplásicos/uso terapéutico , Ceramidas/biosíntesis , Ceramidas/genética , Cistatina A/genética , Células HEK293 , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
9.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 37(2): 109-25, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22561428

RESUMEN

Acid sphingomyelinase (ASM) is one enzyme responsible for the production of ceramide via the hydrolysis of sphingomyelin. Recent findings have revealed the important role of ASM in the initiation of ceramide-induced cell apoptosis, as well as in the pathophysiology of many common diseases (e.g. cardiovascular diseases, diabetes, pulmonary diseases, and neurological diseases). Other studies have also shown that ASM activation may occur through the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), as well as by inflammatory mechanisms that may be induced by environmental and occupational stresses. ASM activation, in turn, may create excess or abnormally distributed ceramides, which could lead to tissue and organ injuries, including to the pulmonary, liver, kidney, and nervous systems. This review will discuss the basic biology of ASM and focus on the role and regulation of ASM in environmental stress responses. We propose that ASM activation is an important factor in environmental health, and that ASM-based therapeutics may have a key role in preventing environmental-induced tissue injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Ceramidas/farmacología , Salud Ambiental , Esfingomielina Fosfodiesterasa/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Ceramidas/metabolismo , Humanos , Hidrólisis , Enfermedades Profesionales/fisiopatología , Especies de Nitrógeno Reactivo/farmacología , Especies Reactivas de Oxígeno/farmacología , Esfingomielinas/química
10.
Stem Cells ; 28(4): 821-31, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20201063

RESUMEN

Niemann-Pick type C (NP-C) disease exhibits neuronal sphingolipid storage and cerebellar Purkinje neuron (PN) loss. Although it is clear that PNs are compromised in this disorder, it remains to be defined how neuronal lipid storage causes the PN loss. Our previous studies have shown that bone marrow-derived mesenchymal stem cells (BM-MSCs) transplantation prevent PN loss in NP-C mice. The aim of the present study was therefore to examine the neuroprotective mechanism of BM-MSCs on PNs. We found that NP-C PNs exhibit abnormal sphingolipid metabolism and defective lysosomal calcium store compared to wild-type mice PNs. BM-MSCs promote the survival of NP-C PNs by correction of the altered calcium homeostasis, restoration of the sphingolipid imbalance, as evidenced by increased sphingosine-1-phosphate levels and decreased sphingosine, and ultimately, inhibition of apoptosis pathways. These effects suggest that BM-MSCs modulate sphingolipid metabolism of endogenous NP-C PNs, resulting in their survival and improved clinical outcome in mice.


Asunto(s)
Lisofosfolípidos/metabolismo , Enfermedades de Niemann-Pick/metabolismo , Células de Purkinje/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Células Madre/metabolismo , Animales , Apoptosis , Células de la Médula Ósea/citología , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Lisosomas/metabolismo , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos BALB C , Enfermedades de Niemann-Pick/patología , Enfermedades de Niemann-Pick/cirugía , Células de Purkinje/citología , Células Madre/citología
11.
FASEB J ; 24(4): 1229-38, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20007509

RESUMEN

A major challenge of assisted reproduction technologies (ARTs) is to mimic the natural environment required to sustain oocyte and embryo survival. Herein, we show that the ceramide-metabolizing enzyme, acid ceramidase (AC), is expressed in human cumulus cells and follicular fluid, essential components of this environment, and that the levels of this enzyme are positively correlated with the quality of human embryos formed in vitro. These observations led us to develop a new approach for oocyte and embryo culture that markedly improved the outcome of in vitro fertilization (IVF). The addition of recombinant AC (rAC) to human and mouse oocyte culture medium maintained their healthy morphology in vitro. Following fertilization, the number of mouse embryos formed in the presence of rAC also was improved (from approximately 40 to 88%), leading to approximately 5-fold more healthy births. To confirm these observations, immature bovine oocytes were matured in vitro and subjected to IVF in the presence of rAC. Significantly more high-grade blastocysts were formed, and the number of morphologically intact, hatched embryos was increased from approximately 24 to 70%. Overall, these data identify AC as an important component of the in vivo oocyte and embryo environment, and provide a novel technology for enhancing the outcome of assisted fertilization. Eliyahu, E., Shtraizent, N., Martinuzzi, K., Barritt, J., He, X., Wei, H., Chaubal, S., Copperman, A. B., Schuchman, E. H. Acid ceramidase improves the quality of oocytes and embryos and the outcome of in vitro fertilization.


Asunto(s)
Ceramidasa Ácida/farmacología , Blastocisto/citología , Células del Cúmulo/citología , Fertilización In Vitro/métodos , Oocitos/citología , Animales , Bovinos , Técnicas de Cultivo de Célula , Técnicas de Cultivo de Embriones , Femenino , Humanos , Masculino , Ratones , Proteínas Recombinantes/farmacología
12.
Front Cell Dev Biol ; 9: 633657, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34026750

RESUMEN

Ceramide is a bioactive signaling lipid involved in the pathogenesis of numerous diseases. It also plays an important role in ischemia reperfusion (IR) injury via activation of inflammatory/oxidative stress-stimulated signaling pathways, resulting in tissue damage. Acid ceramidase is a lipid hydrolase that modulates the levels of ceramide, and as such has a potential therapeutic role in many human diseases where ceramide has been implicated. Here we investigated the therapeutic potential of recombinant acid ceramidase in a murine model of hepatic IR injury. Serum ALT, AST, and LDH activities, as well as oxidative stress (MDA) and inflammatory (MCP-1) markers, were increased in mice subjected to IR compared to a sham group. In contrast, these elevations were significantly lower in an IR group pretreated with a single injection of acid ceramidase. Histological examination by two different assessment criteria also revealed that acid ceramidase pretreatment alleviated IR-induced hepatocyte damage, including reduced evidence of cell death and necrosis. In addition, elevated ceramide and sphingosine levels were observed in the IR group compared to sham, and were markedly reduced when pretreated with acid ceramidase. In contrast, the levels of the protective signaling lipid, sphingosine-1-phosphate (S1P), were reduced following IR and elevated in response to acid ceramidase pretreatment. These changes in sphingolipid levels could be correlated with changes in the activities of several sphingolipid-metabolizing enzymes. Overall, these results indicated that sphingolipid changes were an important pathologic component of hepatic IR injury, and that acid ceramidase administration ameliorated these lipid changes and other downstream pathologic changes.

13.
Mol Med ; 16(7-8): 316-21, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20386867

RESUMEN

Types A and B Niemann-Pick disease (NPD) result from the deficient activity of acid sphingomyelinase (ASM), due to mutations in the sphingomyelin phosphodiesterase 1 (SMPD1) gene. Here we report the identification, characterization and genotype/phenotype correlations of eight novel mutations in six unrelated NPD patients. These mutations included seven missense mutations: c.631T>C (p.W211R), c.757G>C (p.D253H), c.940G>A (p.V314M), c.1280A>G (p.H427R), c.1564A>G (p.N522S), c.1575G>C (p.Q525H) and c.1729A>G (p.H577R), and a novel frameshift mutation, c.1657delACCGCCT (fsT553). Each missense mutation was expressed in 293T or COS-7 cells; mutant enzymes p.W211R, p.D253H, p.H427R and p.H577R had <1% of expressed wild-type activity, whereas p.V314M, p.N522S and p.Q525H had 21.7%, 10.1% and 64% of expressed wild-type activity, respectively. The c.1564A>G mutation obliterated a known N-glycosylation site and its p.N522S mutant enzyme had ~10% of expressed wild-type activity. Western blot analysis revealed that each mutant protein was expressed at near wild-type amounts, despite their differences in residual activity. The novel seven-base deletion occurred at codon 553, leading to a premature truncation after residue 609. The expression studies predicted the clinical phenotypes of the six patients: two type A patients had genotypes with only type A alleles [c.631T>C (p.W211R), c.757G>C (p.D253H) and c.1729A>G (p.H577R)], and the other four type B disease patients had at least one neuroprotective mutant type B allele [c.940G>A (p.V314M), c.1280A>G (p.H427R), c.1564A>G (p.N522S) and c.1575G>C (p.Q525H)] that expressed >5% residual ASM activity. Thus, these new mutations provide novel genotype/phenotype correlations and further document the genetic heterogeneity in types A and B NPD.


Asunto(s)
Mutación del Sistema de Lectura , Mutación Missense , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo B/genética , Esfingomielina Fosfodiesterasa/genética , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , Células COS , Línea Celular , Niño , Preescolar , Chlorocebus aethiops , Clonación Molecular , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Fenotipo , Alineación de Secuencia , Esfingomielina Fosfodiesterasa/metabolismo , Transfección
14.
Cell Physiol Biochem ; 26(4-5): 749-60, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21063112

RESUMEN

BACKGROUND/AIMS: The sphingomyelin/ceramide signaling pathway is an important component of many cellular processes implicated in the pathogenesis of lung disease. Acid sphingomyelinase (ASM) is a key mediator of this pathway, but its specific role in pulmonary fibrosis has not been previously investigated. Here we used the bleomycin model of pulmonary fibrosis to investigate fibrotic responses in normal and ASM knockout (ASM(-/-)) mice, and in NIH3T3 fibroblasts with and without ASM siRNA treatment. METHODS: Mice and cells with and without ASM activity were treated with bleomycin, and the effects on lung inflammation, formation of collagen producing myofibroblasts, and apoptosis were assessed. RESULTS: The development of bleomycin-induced inflammation and fibrosis in wildtype mice correlated with the rapid activation of ASM, and was markedly attenuated in the absence of ASM activity. Along with the elevated ASM activity, there also was an elevation of acid ceramidase (AC) activity, which was sustained for up to 14 days post-bleomycin treatment. Studies in NIH3T3 fibroblasts confirmed these findings, and revealed a direct effect of ASM/AC activation on the formation of myofibroblasts. Cell studies also showed that a downstream effect of bleomycin treatment was the production of sphingosine-1-phosphate. CONCLUSIONS: These data demonstrate that the sphingomyelin/ceramide signaling pathway is involved in the pathogenesis of bleomycin-induced pulmonary fibrosis, and suggest that inhibition of ASM may potentially slow the fibrotic process in the lung.


Asunto(s)
Neumonía/metabolismo , Fibrosis Pulmonar/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Actinas/metabolismo , Animales , Antibióticos Antineoplásicos , Bleomicina , Lisofosfolípidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Neumonía/inducido químicamente , Neumonía/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/genética , Esfingosina/análogos & derivados , Esfingosina/metabolismo
15.
Front Pharmacol ; 11: 594471, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33192534

RESUMEN

[This corrects the article DOI: 10.3389/fphar.2018.01504.].

16.
Sci Transl Med ; 11(506)2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31434754

RESUMEN

Niemann-Pick disease type A (NPD-A) is a lysosomal storage disorder characterized by neurodegeneration and early death. It is caused by loss-of-function mutations in the gene encoding for acid sphingomyelinase (ASM), which hydrolyzes sphingomyelin into ceramide. Here, we evaluated the safety of cerebellomedullary (CM) cistern injection of adeno-associated viral vector serotype 9 encoding human ASM (AAV9-hASM) in nonhuman primates (NHP). We also evaluated its therapeutic benefit in a mouse model of the disease (ASM-KO mice). We found that CM injection in NHP resulted in widespread transgene expression within brain and spinal cord cells without signs of toxicity. CM injection in the ASM-KO mouse model resulted in hASM expression in cerebrospinal fluid and in different brain areas without triggering an inflammatory response. In contrast, direct cerebellar injection of AAV9-hASM triggered immune response. We also identified a minimally effective therapeutic dose for CM injection of AAV9-hASM in mice. Two months after administration, the treatment prevented motor and memory impairment, sphingomyelin (SM) accumulation, lysosomal enlargement, and neuronal death in ASM-KO mice. ASM activity was also detected in plasma from AAV9-hASM CM-injected ASM-KO mice, along with reduced SM amount and decreased inflammation in the liver. Our results support CM injection for future AAV9-based clinical trials in NPD-A as well as other lysosomal storage brain disorders.


Asunto(s)
Dependovirus/metabolismo , Terapia Genética , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/terapia , Serogrupo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Humanos , Inflamación/patología , Inyecciones , Hígado/patología , Ratones Noqueados , Actividad Motora , Primates , Esfingomielina Fosfodiesterasa/administración & dosificación , Esfingomielina Fosfodiesterasa/sangre , Esfingomielina Fosfodiesterasa/genética , Transgenes
17.
Mol Genet Metab ; 95(3): 152-62, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18815062

RESUMEN

Herein we describe detailed characterization of four common mutations (L302P, H421Y, R496L and DeltaR608) within the acid sphingomyelinase (ASM) gene causing types A and B Niemann-Pick disease (NPD). In vitro and in situ enzyme assays revealed marked deficiencies of ASM activity in NPD cell lines homoallelic for each mutation, although Western blotting and fluorescent microscopy showed that the mutant ASM polypeptides were expressed at normal levels and trafficked to lysosomes. Co-immunoprecipitation of the polypeptides with the ER chaperone, BiP, confirmed these findings, as did in vitro expression of the mutant cDNAs in reticulocyte lysates. We further developed a computer assisted, three-dimensional model of human ASM based on homologies to known proteins, and used this model to map each NPD mutation in relation to putative substrate binding, hydrolysis and zinc-binding domains. Lastly, we generated transgenic mice expressing the R496L and DeltaR608 mutations on the complete ASM knock-out background (ASMKO), and established breeding colonies for the future evaluation of enzyme enhancement therapies. Analysis of these mice demonstrated that the mutant ASM transgenes were expressed at high levels in the brain, and in the case of the DeltaR608 mutation, produced residual ASM activity that was significantly above the ASMKO background.


Asunto(s)
Mutación , Enfermedades de Niemann-Pick/enzimología , Enfermedades de Niemann-Pick/genética , Esfingomielina Fosfodiesterasa/genética , Secuencia de Aminoácidos , Animales , Encéfalo/enzimología , Línea Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Transporte de Proteínas , Alineación de Secuencia , Esfingomielina Fosfodiesterasa/química , Esfingomielina Fosfodiesterasa/metabolismo
18.
FASEB J ; 21(7): 1403-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17264167

RESUMEN

Recent studies suggest that the lipid, ceramide, induces the default apoptosis process in eggs. Yet, it is obscure how newly formed embryos overcome this fate. Acid ceramidase (AC) is a key regulatory enzyme involved in ceramide metabolism, and mutations in the AC gene (Asah1) result in Farber Lipogranulomatosis, a fatal human genetic disorder. Our previous studies revealed that AC knockout (Asah1-/-) mice had a lethal phenotype, and herein we reveal the mechanism underlying this observation. A single-cell, polymerase chain reaction (PCR) genotyping method was developed to analyze individual embryos from Asah1 +/- intercrosses. Combined with Annexin V staining, this genotype analysis demonstrated that Asah1-/- embryos could not survive beyond the 2-cell stage, and underwent apoptotic death. Notably, sphingosine-1-phosphate (S1P) treatment of early 2-cell embryos from the Asah1 +/- intercrosses rescued Asah1-/- embryos, and enabled their progression from the 2-cell to 4-8-cell stage. Quantitative PCR also revealed that expression of the Asah1 gene in healthy embryos was initiated at the 2-cell stage, coincident with embryonic genome activation (EGA). AC activity and Western blot analyses further demonstrated high expression and activity of the enzyme in normal, unfertilized eggs, which likely provide the protein to newly formed embryos prior to EGA. Based on these observations, we suggest that AC is an essential factor required for embryo survival that functions by removing ceramide from the newly formed embryos, thus inhibiting the default apoptosis pathway.


Asunto(s)
Desarrollo Embrionario/fisiología , Galactosilgalactosilglucosilceramidasa/fisiología , Animales , Apoptosis , Secuencia de Bases , Western Blotting , Cartilla de ADN , Embrión de Mamíferos/citología , Femenino , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética
19.
Front Pharmacol ; 9: 1504, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30687087

RESUMEN

Tissue fibrosis, including pulmonary fibrosis, hepatic fibrosis, and cardiac fibrosis, is an important stage in the development of many diseases. It can lead to structural damage and dysfunction and even severe carcinogenesis or death. There is currently no effective method for the treatment of fibrosis. At present, the molecular mechanism of tissue fibrosis has not yet been fully elucidated, but many studies have demonstrated that it is involved in conveying the complex messages between fibroblasts and various cytokines. Sphingosine 1-phosphate (S1P) is a naturally bioactive sphingolipid. S1P and the related signaling pathways are important intracellular metabolic pathways involved in many life activities, including cell proliferation, differentiation, apoptosis, and cellular signal transduction. Increasing evidence suggests that S1P and its signaling pathways play an important role in the development of tissue fibrosis; however, the mechanisms of these effects have not yet been fully elucidated, and even the role of S1P and its signaling pathways are still controversial. This article focuses on the role of S1P and the related signaling pathways in the development of fibrosis of lung, liver, heart, and other tissues, with emphasis on the application of inhibitors of some of molecules in the pathway in clinical treatment of fibrosis diseases.

20.
J Lipids ; 2018: 3646725, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29610685

RESUMEN

Ceramide, a bioactive membrane sphingolipid, functions as an important second messenger in apoptosis and cell signaling. In response to stresses, it may be generated by de novo synthesis, sphingomyelin hydrolysis, and/or recycling of complex sphingolipids. It is cleared from cells through the activity of ceramidases, phosphorylation to ceramide-1-phosphate, or resynthesis into more complex sphingolipids. Ischemia/reperfusion (IR) injury occurs when oxygen/nutrition is rapidly reintroduced into ischemic tissue, resulting in cell death and tissue damage, and is a major concern in diverse clinical settings, including organ resection and transplantation. Numerous reports show that ceramide levels are markedly elevated during IR. Mitochondria are major sites of reactive oxygen species (ROS) production and play a key role in IR-induced and ceramide-mediated cell death and tissue damage. During the development of IR injury, the initial response of ROS and TNF-alpha production activates two major ceramide generating pathways (sphingomyelin hydrolysis and de novo ceramide synthesis). The increased ceramide has broad effects depending on the IR phases, including both pro- and antiapoptotic effects. Therefore, strategies that reduce the levels of ceramide, for example, by modulation of ceramidase and/or sphingomyelinases activities, may represent novel and promising therapeutic approaches to prevent or treat IR injury in diverse clinical settings.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA