Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Autophagy ; 17(8): 1947-1961, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-32835606

RESUMO

1-Deoxysphingolipids (deoxySLs) are atypical sphingolipids of clinical relevance as they are elevated in plasma of patients suffering from hereditary sensory and autonomic neuropathy (HSAN1) or type 2 diabetes. Their neurotoxicity is described best but they inflict damage to various cell types by an uncertain pathomechanism. Using mouse embryonic fibroblasts and an alkyne analog of 1-deoxysphinganine (doxSA), the metabolic precursor of all deoxySLs, we here study the impact of deoxySLs on macroautophagy/autophagy, the regulated degradation of dysfunctional or expendable cellular components. We find that deoxySLs induce autophagosome and lysosome accumulation indicative of an increase in autophagic flux. The autophagosomal machinery targets damaged mitochondria that have accumulated N-acylated doxSA metabolites, presumably deoxyceramide and deoxydihydroceramide, and show aberrant swelling and tubule formation. Autophagosomes and lysosomes also interact with cellular lipid aggregates and crystals that occur upon cellular uptake and N-acylation of monomeric doxSA. As crystals entering the lysophagosomal apparatus in phagocytes are known to trigger the NLRP3 inflammasome, we also treated macrophages with doxSA. We demonstrate the activation of the NLRP3 inflammasome by doxSLs, prompting the release of IL1B from primary macrophages. Taken together, our data establish an impact of doxSLs on autophagy and link doxSL pathophysiology to inflammation and the innate immune system.Abbreviations: alkyne-doxSA: (2S,3R)-2-aminooctadec-17yn-3-ol; alkyne-SA: (2S,3R)-2- aminooctadec-17yn-1,3-diol; aSA: alkyne-sphinganine; ASTM-BODIPY: azido-sulfo-tetramethyl-BODIPY; CerS: ceramide synthase; CMR: clonal macrophage reporter; deoxySLs: 1-deoxysphingolipids; dox(DH)Cer: 1-deoxydihydroceramide; doxCer: 1-deoxyceramide; doxSA: 1-deoxysphinganine; FB1: fumonisin B1; HSAN1: hereditary sensory and autonomic neuropathy type 1; LC3: MAP1LC3A and MAP1LC3B; LPS: lipopolysaccharide; MEF: mouse embryonal fibroblasts; MS: mass spectrometry; N3635P: azido-STAR635P; N3Cy3: azido-cyanine 3; N3picCy3: azido-picolylcyanine 3; NLRP3: NOD-like receptor pyrin domain containing protein 3; P4HB: prolyl 4-hydroxylase subunit beta; PINK1: PTEN induced putative kinase 1; PYCARD/ASC: PYD and CARD domain containing; SPTLC1: serine palmitoyltransferase long chain base subunit 1; SQSTM1: sequestosome 1; TLC: thin layer chromatography.


Assuntos
Autofagossomos/efeitos dos fármacos , Inflamassomos/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/efeitos dos fármacos , Esfingolipídeos/farmacologia , Animais , Autofagossomos/metabolismo , Autofagia/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Inflamassomos/metabolismo , Inflamação/metabolismo , Lisossomos/metabolismo , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
2.
J Cereb Blood Flow Metab ; 40(6): 1338-1350, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31357902

RESUMO

In Tay-Sachs and Sandhoff disease, a deficiency of the lysosomal enzyme ß-hexosaminidase causes GM2 and other gangliosides to accumulate in neurons and triggers neurodegeneration. Although the pathology centers on neurons, ß-hexosaminidase is mainly expressed outside of neurons, suggesting that gene therapy of these diseases should target non-neuronal cells to reconstitute physiological conditions. Here, we tested in Hexb-/- mice, a model of Sandhoff disease, to determine whether endothelial expression of the genes for human ß-hexosaminidase subunit A and B (HEXA, HEXB) is able to reduce disease symptoms and prolong survival of the affected mice. The brain endothelial selective vectors AAV-BR1-CAG-HEXA and AAV-BR1-CAG-HEXB transduced brain endothelial cells, which subsequently released ß-hexosaminidase enzyme. In vivo intravenous administration of the gene vectors to adult and neonatal mice prolonged survival. They improved neurological function and reduced accumulation of the ganglioside GM2 and the glycolipid GA2 as well as astrocytic activation. Overall, the data demonstrate that endothelial cells are a suitable target for intravenous gene therapy of GM2 gangliosidoses and possibly other lysosomal storage disorders.


Assuntos
Células Endoteliais , Terapia Genética/métodos , Doença de Sandhoff , Cadeia alfa da beta-Hexosaminidase/administração & dosagem , Cadeia beta da beta-Hexosaminidase/administração & dosagem , Animais , Encéfalo , Dependovirus , Modelos Animais de Doenças , Vetores Genéticos , Humanos , Camundongos , Camundongos Knockout , Transdução Genética , Cadeia alfa da beta-Hexosaminidase/genética , Cadeia beta da beta-Hexosaminidase/genética
3.
J Biol Chem ; 292(15): 6177-6189, 2017 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-28258214

RESUMO

The lysosomal acid ß-glucosidase GBA1 and the non-lysosomal ß-glucosidase GBA2 degrade glucosylceramide (GlcCer) to glucose and ceramide in different cellular compartments. Loss of GBA2 activity and the resulting accumulation of GlcCer results in male infertility, whereas mutations in the GBA1 gene and loss of GBA1 activity cause the lipid-storage disorder Gaucher disease. However, the role of GBA2 in Gaucher disease pathology and its relationship to GBA1 is not well understood. Here, we report a GBA1-dependent down-regulation of GBA2 activity in patients with Gaucher disease. Using an experimental approach combining cell biology, biochemistry, and mass spectrometry, we show that sphingosine, the cytotoxic metabolite accumulating in Gaucher cells through the action of GBA2, directly binds to GBA2 and inhibits its activity. We propose a negative feedback loop, in which sphingosine inhibits GBA2 activity in Gaucher cells, preventing further sphingosine accumulation and, thereby, cytotoxicity. Our findings add a new chapter to the understanding of the complex molecular mechanism underlying Gaucher disease and the regulation of ß-glucosidase activity in general.


Assuntos
Regulação para Baixo , Doença de Gaucher/enzimologia , Regulação Enzimológica da Expressão Gênica , Modelos Biológicos , Esfingosina/metabolismo , beta-Glucosidase/biossíntese , Animais , Linhagem Celular , Doença de Gaucher/genética , Glucosilceramidase , Glucosilceramidas/genética , Glucosilceramidas/metabolismo , Humanos , Masculino , Camundongos , Esfingosina/genética , beta-Glucosidase/genética
4.
J Lipid Res ; 58(1): 42-59, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27881717

RESUMO

1-Deoxysphingolipids (deoxySLs) are atypical sphingolipids that are elevated in the plasma of patients with type 2 diabetes and hereditary sensory and autonomic neuropathy type 1 (HSAN1). Clinically, diabetic neuropathy and HSAN1 are very similar, suggesting the involvement of deoxySLs in the pathology of both diseases. However, very little is known about the biology of these lipids and the underlying pathomechanism. We synthesized an alkyne analog of 1-deoxysphinganine (doxSA), the metabolic precursor of all deoxySLs, to trace the metabolism and localization of deoxySLs. Our results indicate that the metabolism of these lipids is restricted to only some lipid species and that they are not converted to canonical sphingolipids or fatty acids. Furthermore, exogenously added alkyne-doxSA [(2S,3R)-2-aminooctadec-17-yn-3-ol] localized to mitochondria, causing mitochondrial fragmentation and dysfunction. The induced mitochondrial toxicity was also shown for natural doxSA, but not for sphinganine, and was rescued by inhibition of ceramide synthase activity. Our findings therefore indicate that mitochondrial enrichment of an N-acylated doxSA metabolite may contribute to the neurotoxicity seen in diabetic neuropathy and HSAN1. Hence, we provide a potential explanation for the characteristic vulnerability of peripheral nerves to elevated levels of deoxySLs.


Assuntos
Diabetes Mellitus Tipo 2/sangue , Neuropatias Diabéticas/sangue , Neuropatias Hereditárias Sensoriais e Autônomas/sangue , Esfingolipídeos/sangue , Animais , Diabetes Mellitus Tipo 2/patologia , Neuropatias Diabéticas/patologia , Neuropatias Hereditárias Sensoriais e Autônomas/patologia , Humanos , Lipídeos/sangue , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Oxirredutases/metabolismo , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Esfingolipídeos/síntese química , Esfingolipídeos/farmacologia
5.
J Lipid Res ; 58(1): 60-71, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27872144

RESUMO

The 1-deoxysphingolipids (1-deoxySLs) are atypical sphingolipids (SLs) that are formed when serine palmitoyltransferase condenses palmitoyl-CoA with alanine instead of serine during SL synthesis. The 1-deoxySLs are toxic to neurons and pancreatic ß-cells. Pathologically elevated 1-deoxySLs cause the inherited neuropathy, hereditary sensory autonomic neuropathy type 1 (HSAN1), and are also found in T2D. Diabetic sensory polyneuropathy (DSN) and HSAN1 are clinically very similar, suggesting that 1-deoxySLs may be implicated in both pathologies. The 1-deoxySLs are considered to be dead-end metabolites, as they lack the C1-hydroxyl group, which is essential for the canonical degradation of SLs. Here, we report a previously unknown metabolic pathway, which is capable of degrading 1-deoxySLs. Using a variety of metabolic labeling approaches and high-resolution high-accuracy MS, we identified eight 1-deoxySL downstream metabolites, which appear to be formed by cytochrome P450 (CYP)4F enzymes. Comprehensive inhibition and induction of CYP4F enzymes blocked and stimulated, respectively, the formation of the downstream metabolites. Consequently, CYP4F enzymes might be novel therapeutic targets for the treatment of HSAN1 and DSN, as well as for the prevention of T2D.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Neuropatias Diabéticas/metabolismo , Neuropatias Hereditárias Sensoriais e Autônomas/metabolismo , Esfingolipídeos/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/genética , Neuropatias Diabéticas/genética , Neuropatias Diabéticas/patologia , Neuropatias Hereditárias Sensoriais e Autônomas/patologia , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Redes e Vias Metabólicas/genética , Camundongos , Mutação , Oxirredução , Serina C-Palmitoiltransferase/metabolismo
6.
J Lipid Res ; 57(10): 1934-1947, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27565170

RESUMO

The demand to study the cellular localization of specific lipids has led to recent advances in lipid probes and microscopy. Alkyne lipids bear a small, noninterfering tag and can be detected upon click reaction with an azide-coupled reporter. Fluorescent alkyne lipid imaging crucially depends on appropriate azide reporters and labeling protocols that allow for an efficient click reaction and therefore a sensitive detection. We synthesized several azide reporters with different spacer components and tested their suitability for alkyne lipid imaging in fixed cells. The implementation of a copper-chelating picolyl moiety into fluorescent or biotin-based azide reagents strongly increased the sensitivity of the imaging routine. We demonstrate the applicability and evaluate the performance of this approach using different lipid classes and experimental setups. As azide picolyl reporters allow for reduced copper catalyst concentrations, they also enable coimaging of alkyne lipids with multiple fluorescent proteins including enhanced green fluorescent protein. Alternatively, and as we also show, microscopy of alkyne lipids can be combined with protein detection by immunocytochemistry. In summary, we present a robust, sensitive, and highly versatile protocol for the labeling of alkyne lipids with azide-coupled reporters for fluorescence microscopy that can be combined with different protein detection and imaging techniques.


Assuntos
Corantes Fluorescentes/química , Proteínas de Fluorescência Verde/química , Lipídeos/química , Coloração e Rotulagem/métodos , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microscopia de Fluorescência/métodos
7.
Neuromolecular Med ; 17(1): 47-57, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25567748

RESUMO

1-Deoxysphingolipids (1-deoxySL) are atypical sphingolipids that are formed by the enzyme serine palmitoyltransferase (SPT) due to a promiscuous use of L-alanine over its canonical substrate L-serine. Several mutations in SPT are associated with the hereditary sensory and autonomic neuropathy type I (HSAN1). The current hypothesis is that these mutations induce a permanent shift in the affinity from L-serine toward L-alanine which results in a pathologically increased 1-deoxySL formation in HSAN1 patients. Also, wild-type SPT forms 1-deoxySL under certain conditions, and elevated levels were found in individuals with the metabolic syndrome and diabetes. However, the molecular mechanisms which control the substrate shift of the wild-type enzyme are not understood. Here, we report a novel SPTLC2-S384F variant in two unrelated HSAN1 families. Affected patients showed elevated plasma 1-deoxySL levels and expression of the S384F mutant in HEK293 cells increased 1-deoxySL formation. Previously, S384 has been reported as one of the two (S384 and Y387) putative phosphorylation sites in SPTLC2. The phosphorylation of wild-type SPTLC2 was confirmed by isoelectric focusing. The impact of an S384 phosphorylation on SPT activity was tested by creating mutants mimicking either a constitutively phosphorylated (S384D, S384E) or non-phosphorylated (S384A, Y387F, Y387F+S384A) protein. The S384D but not the S384E variant was associated with increased 1-deoxySL formation. The other mutations had no influence on activity and substrate affinity. In summary, our data show that S384F is a novel mutation in HSAN1 and that the substrate specificity of wild-type SPT might by dynamically regulated by a phosphorylation at this position.


Assuntos
Neuropatias Hereditárias Sensoriais e Autônomas/genética , Serina C-Palmitoiltransferase/genética , Idoso , Sequência de Aminoácidos , Substituição de Aminoácidos , Sequência Conservada , Eletroforese em Gel Bidimensional , Feminino , Células HEK293 , Heterozigoto , Homozigoto , Humanos , Focalização Isoelétrica , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Dados de Sequência Molecular , Condução Nervosa , Linhagem , Fosforilação , Fosfosserina/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Serina C-Palmitoiltransferase/química , Serina C-Palmitoiltransferase/fisiologia , Especificidade da Espécie , Esfingolipídeos/metabolismo , Especificidade por Substrato
8.
Biochim Biophys Acta ; 1831(3): 589-94, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23246574

RESUMO

Lipid droplets are ubiquitous cellular organelles that allow cells to store large amounts of neutral lipids for membrane synthesis and energy supply in times of starvation. Compared to other cellular organelles, lipid droplets are structurally unique as they are made of a hydrophobic core of neutral lipids and are separated to the cytosol only by a surrounding phospholipid monolayer. This phospholipid monolayer consists of over a hundred different phospholipid molecular species of which phosphatidylcholine is the most abundant lipid class. However, lipid droplets lack some indispensable activities of the phosphatidylcholine biogenic pathways suggesting that they partially depend on other organelles for phosphatidylcholine synthesis. Here, we discuss very recent data on the composition, origin, transport and function of the phospholipid monolayer with a particular emphasis on the phosphatidylcholine metabolism on and for lipid droplets. In addition, we highlight two very important quantitative aspects: (i) The amount of phospholipid required for lipid droplet monolayer expansion is remarkably small and (ii) to maintain the invariably round shape of lipid droplets, a cell must have a highly sensitive but so far unknown mechanism that regulates the ratio of phospholipid to neutral lipid in lipid droplets. This article is part of a Special Issue entitled Phospholipids and Phospholipid Metabolism.


Assuntos
Vesículas Citoplasmáticas/metabolismo , Metabolismo dos Lipídeos , Fosfatidilcolinas/metabolismo , Animais , Transporte Biológico , Vesículas Citoplasmáticas/química , Humanos , Forma das Organelas , Fosfatidilcolinas/química , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo
9.
J Clin Invest ; 121(12): 4735-45, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22045570

RESUMO

Hereditary sensory and autonomic neuropathy type 1 (HSAN1) causes sensory loss that predominantly affects the lower limbs, often preceded by hyperpathia and spontaneous shooting or lancinating pain. It is caused by several missense mutations in the genes encoding 2 of the 3 subunits of the enzyme serine palmitoyltransferase (SPT). The mutant forms of the enzyme show a shift from their canonical substrate L-serine to the alternative substrate L-alanine. This shift leads to increased formation of neurotoxic deoxysphingolipids (dSLs). Our initial analysis showed that in HEK cells transfected with SPTLC1 mutants, dSL generation was modulated in vitro in the presence of various amino acids. We therefore examined whether in vivo specific amino acid substrate supplementation influenced dSL levels and disease severity in HSAN1. In mice bearing a transgene expressing the C133W SPTLC1 mutant linked to HSAN1, a 10% L-serine­enriched diet reduced dSL levels. L-serine supplementation also improved measures of motor and sensory performance as well as measures of male fertility. In contrast, a 10% L-alanine­enriched diet increased dSL levels and led to severe peripheral neuropathy. In a pilot study with 14 HSAN1 patients, L-serine supplementation similarly reduced dSL levels. These observations support the hypothesis that an altered substrate selectivity of the mutant SPT is key to the pathophysiology of HSAN1 and raise the prospect of l-serine supplementation as a first treatment option for this disorder.


Assuntos
Neuropatias Hereditárias Sensoriais e Autônomas/tratamento farmacológico , Neurotoxinas/biossíntese , Serina/uso terapêutico , Esfingosina/análogos & derivados , Administração Oral , Adulto , Idoso , Alanina/toxicidade , Animais , Depressão Química , Relação Dose-Resposta a Droga , Feminino , Neuropatias Hereditárias Sensoriais e Autônomas/genética , Neuropatias Hereditárias Sensoriais e Autônomas/metabolismo , Humanos , Infertilidade Masculina/tratamento farmacológico , Infertilidade Masculina/genética , Lipídeos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Percepção da Dor/efeitos dos fármacos , Projetos Piloto , Mutação Puntual , Desempenho Psicomotor/efeitos dos fármacos , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/patologia , Serina/administração & dosagem , Serina/química , Serina C-Palmitoiltransferase/deficiência , Serina C-Palmitoiltransferase/genética , Esfingolipídeos/metabolismo , Esfingosina/biossíntese , Estereoisomerismo , Adulto Jovem
10.
Hum Mutat ; 32(6): E2211-25, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21618344

RESUMO

Hereditary sensory and autonomic neuropathy type I (HSAN-I) is an axonal peripheral neuropathy leading to progressive distal sensory loss and severe ulcerations. Mutations in SPTLC1 and SPTLC2, encoding the two subunits of serine palmitoyltransferase (SPT), the enzyme catalyzing the first and rate-limiting step in the de novo synthesis of sphingolipids, have been reported to cause HSAN-I. Here, we demonstrate that the SPTLC1 mutations p.S331F and p.A352V result in a reduction of SPT activity in vitro and are associated with increased levels of the deoxysphingoid bases 1-deoxy-sphinganine and 1-deoxymethyl-sphinganine in patients' plasma samples. Stably expressing p.S331F-SPTLC1 HEK293T cell lines likewise show accumulation of deoxysphingoid bases, but this accumulation is not observed in HEK293T cells overexpressing p.A352V-SPTLC1. These results confirm that the increased formation of deoxysphingoid bases is a key feature for HSAN-I as it is associated with all pathogenic SPTLC1 and SPTLC2 mutations reported so far, but also warrant for caution in the interpretation of in vitro data.


Assuntos
Neuropatias Hereditárias Sensoriais e Autônomas/genética , Mutação , Serina C-Palmitoiltransferase/genética , Expressão Gênica , Células HEK293 , Neuropatias Hereditárias Sensoriais e Autônomas/sangue , Neuropatias Hereditárias Sensoriais e Autônomas/patologia , Humanos , Lipídeos , Conformação Proteica , Serina C-Palmitoiltransferase/química , Esfingosina/análogos & derivados , Esfingosina/sangue
11.
Am J Hum Genet ; 87(4): 513-22, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20920666

RESUMO

Hereditary sensory and autonomic neuropathy type I (HSAN-I) is an axonal peripheral neuropathy associated with progressive distal sensory loss and severe ulcerations. Mutations in the first subunit of the enzyme serine palmitoyltransferase (SPT) have been associated with HSAN-I. The SPT enzyme catalyzes the first and rate-limiting step in the de novo sphingolipid synthesis pathway. However, different studies suggest the implication of other genes in the pathology of HSAN-I. Therefore, we screened the two other known subunits of SPT, SPTLC2 and SPTLC3, in a cohort of 78 HSAN patients. No mutations were found in SPTLC3, but we identified three heterozygous missense mutations in the SPTLC2 subunit of SPT in four families presenting with a typical HSAN-I phenotype. We demonstrate that these mutations result in a partial to complete loss of SPT activity in vitro and in vivo. Moreover, they cause the accumulation of the atypical and neurotoxic sphingoid metabolite 1-deoxy-sphinganine. Our findings extend the genetic heterogeneity in HSAN-I and enlarge the group of HSAN neuropathies associated with SPT defects. We further show that HSAN-I is consistently associated with an increased formation of the neurotoxic 1-deoxysphinganine, suggesting a common pathomechanism for HSAN-I.


Assuntos
Neuropatias Hereditárias Sensoriais e Autônomas/genética , Serina C-Palmitoiltransferase/genética , Sequência de Bases , Linhagem Celular , Clonagem Molecular , Estudos de Coortes , Análise Mutacional de DNA , Primers do DNA/genética , Teste de Complementação Genética , Humanos , Repetições de Microssatélites/genética , Dados de Sequência Molecular , Mutação de Sentido Incorreto/genética , Serina C-Palmitoiltransferase/metabolismo
12.
J Biol Chem ; 285(15): 11178-87, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20097765

RESUMO

HSAN1 is an inherited neuropathy found to be associated with several missense mutations in the SPTLC1 subunit of serine palmitoyltransferase (SPT). SPT catalyzes the condensation of serine and palmitoyl-CoA, the initial step in the de novo synthesis of sphingolipids. Here we show that the HSAN1 mutations induce a shift in the substrate specificity of SPT, which leads to the formation of the two atypical deoxy-sphingoid bases (DSBs) 1-deoxy-sphinganine and 1-deoxymethyl-sphinganine. Both metabolites lack the C(1) hydroxyl group of sphinganine and can therefore neither be converted to complex sphingolipids nor degraded. Consequently, they accumulate in the cell, as demonstrated in HEK293 cells overexpressing mutant SPTLC1 and lymphoblasts of HSAN1 patients. Elevated DSB levels were also found in the plasma of HSAN1 patients and confirmed in three groups of HSAN1 patients with different SPTLC1 mutations. The DSBs show pronounced neurotoxic effects on neurite formation in cultured sensory neurons. The neurotoxicity co-occurs with a disturbed neurofilament structure in neurites when cultured in the presence of DSBs. Based on these observations, we conclude that HSAN1 is caused by a gain of function mutation, which results in the formation of two atypical and neurotoxic sphingolipid metabolites.


Assuntos
Neuropatias Hereditárias Sensoriais e Autônomas/genética , Neurotoxinas/metabolismo , Serina C-Palmitoiltransferase/genética , Esfingolipídeos/metabolismo , Animais , Embrião de Galinha , Fumonisinas/química , Neuropatias Hereditárias Sensoriais e Autônomas/etiologia , Humanos , Lipídeos/química , Linfócitos/citologia , Modelos Biológicos , Mutação , Neuritos/metabolismo , Neurônios/metabolismo , Serina C-Palmitoiltransferase/fisiologia , Especificidade por Substrato
13.
J Neurosci ; 29(46): 14646-51, 2009 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19923297

RESUMO

Mutations in the SPTLC1 subunit of serine palmitoyltransferase (SPT) cause an adult-onset, hereditary sensory, and autonomic neuropathy type I (HSAN1). We previously reported that mice bearing a transgene-expressing mutant SPTLC1 (tgSPTLC1(C133W)) show a reduction in SPT activity and hyperpathia at 10 months of age. Now analyzed at a later age, we find these mice develop sensory loss with a distal small fiber neuropathy and peripheral myelinopathy. This phenotype is largely reversed when these mice are crossed with transgenic mice overexpressing wild-type SPTLC1 showing that the mutant SPTLC1 protein is not inherently toxic. Simple loss of SPT activity also cannot account for the HSAN1 phenotype, since heterozygous SPTLC1 knock-out mice have reduced SPT activity but are otherwise normal. Rather, the presence of two newly identified, potentially deleterious deoxysphingoid bases in the tgSPTLC1(C133W), but not in the wild-type, double-transgenic tgSPTLC1(WT + C133W) or SPTLC1(+/-) mice, suggests that the HSAN1 mutations alter amino acid selectivity of the SPT enzyme such that palmitate is condensed with alanine and glycine, in addition to serine. This observation is consistent with the hypothesis that HSAN1 is the result of a gain-of-function mutation in SPTLC1 that leads to accumulation of a toxic metabolite.


Assuntos
Expressão Gênica , Neuropatias Hereditárias Sensoriais e Autônomas/genética , Fenótipo , Subunidades Proteicas/genética , Serina C-Palmitoiltransferase/genética , Esfingolipídeos/metabolismo , Animais , Cricetinae , Neuropatias Hereditárias Sensoriais e Autônomas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Camundongos Transgênicos , Subunidades Proteicas/biossíntese , Subunidades Proteicas/fisiologia , Serina C-Palmitoiltransferase/biossíntese , Serina C-Palmitoiltransferase/fisiologia , Esfingolipídeos/toxicidade
14.
J Biol Chem ; 284(39): 26322-30, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19648650

RESUMO

The enzyme serine palmitoyltransferase (SPT) catalyzes the rate-limiting step in the de novo synthesis of sphingolipids. Previously the mammalian SPT was described as a heterodimer composed of two subunits, SPTLC1 and SPTLC2. Recently we identified a novel third SPT subunit (SPTLC3). SPTLC3 shows about 68% identity to SPTLC2 and also includes a pyridoxal phosphate consensus motif. Here we report that the overexpression of SPTLC3 in HEK293 cells leads to the formation of two new sphingoid base metabolites, namely C(16)-sphinganine and C(16)-sphingosine. SPTLC3-expressing cells have higher in vitro SPT activities with lauryl- and myristoyl-CoA than SPTLC2-expressing cells, and SPTLC3 mRNA expression levels correlate closely with the C(16)-sphinganine synthesis rates in various human and murine cell lines. Approximately 15% of the total sphingolipids in human plasma contain a C(16) backbone and are found in the high density and low density but not the very low density lipoprotein fraction. In conclusion, we show that the SPTLC3 subunit generates C(16)-sphingoid bases and that sphingolipids with a C(16) backbone constitute a significant proportion of human plasma sphingolipids.


Assuntos
Serina C-Palmitoiltransferase/metabolismo , Esfingolipídeos/metabolismo , Catálise/efeitos dos fármacos , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Expressão Gênica , Humanos , Cinética , Espectrometria de Massas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina/farmacologia , Serina C-Palmitoiltransferase/sangue , Serina C-Palmitoiltransferase/genética , Esfingolipídeos/sangue , Esfingolipídeos/química , Esfingosina/análogos & derivados , Esfingosina/química , Esfingosina/metabolismo
15.
J Lipid Res ; 50(6): 1237-44, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19181628

RESUMO

Serine palmitoyltransferase (SPT) catalyzes the condensation of l-serine and palmitoyl-CoA, which is the rate-limiting step in the de novo synthesis of sphingolipids. SPT activity is commonly measured by monitoring the incorporation of radiolabeled l-serine into 3-ketodihydrosphingosine. In this article, we introduce several adaptations of the established protocol to improve sensitivity, reproducibility, and practicability of the assay. A significant improvement of this new protocol is the possibility to measure SPT activity in total cell lysate instead of microsomes. The assay is furthermore extended by the introduction of a nonradioactive, HPLC-based detection protocol. The suggested HPLC method offers several advantages, most importantly, a 20-fold lower detection limit compared with the radioactive assay and the possibility to use an internal standard to correct for variation in the extraction.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Serina C-Palmitoiltransferase/análise , Radioisótopos de Carbono , Linhagem Celular , Cromatografia Líquida de Alta Pressão/normas , Cromatografia Líquida de Alta Pressão/estatística & dados numéricos , Ditiotreitol/farmacologia , Humanos , Cinética , Palmitoil Coenzima A/metabolismo , Palmitoil-CoA Hidrolase/metabolismo , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Serina C-Palmitoiltransferase/genética , Serina C-Palmitoiltransferase/metabolismo , Sphingomonas/enzimologia , Sphingomonas/genética , Especificidade por Substrato
16.
Neurogenetics ; 10(2): 135-43, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19132419

RESUMO

Hereditary sensory neuropathy type 1 (HSAN I) is an autosomal dominant inherited neurodegenerative disorder of the peripheral nervous system associated with mutations in the SPTLC1 subunit of the serine palmitoyltransferase (SPT). Four missense mutations (C133W, C133Y, V144D and G387A) in SPTLC1 were reported to cause HSAN I. SPT catalyses the condensation of Serine and Palmitoyl-CoA, which is the first and rate-limiting step in the de novo synthesis of ceramides. Earlier studies showed that C133W and C133Y mutants have a reduced activity, whereas the impact of the V144D and G387A mutations on the human enzyme was not tested yet. In this paper, we show that none of the HSAN I mutations interferes with SPT complex formation. We demonstrate that also V144D has a reduced SPT activity, however to a lower extent than C133W and C133Y. In contrast, the G387A mutation showed no influence on SPT activity. Furthermore, the growth phenotype of LY-B cells--a SPTLC1 deficient CHO cell line--could be reversed by expressing either the wild-type SPTLC1 or the G387A mutant, but not the C133W mutant. This indicates that the G387A mutation is most likely not directly associated with HSAN I. These findings were genetically confirmed by the identification of a nuclear HSAN family which showed segregation of the G387A variant as a non-synonymous SNP.


Assuntos
Análise Mutacional de DNA , Neuropatias Hereditárias Sensoriais e Autônomas/genética , Mutação de Sentido Incorreto , Serina C-Palmitoiltransferase/genética , Animais , Linhagem Celular , Feminino , Humanos , Lactente , Masculino , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA