Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
J Lipid Res ; : 100587, 2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-38950680

RESUMO

Lipotoxicity has been considered the main cause of pancreatic beta-cell failure during type 2 diabetes development. Lipid droplets (LD) are believed to regulate the beta-cell sensitivity to free fatty acids (FFA), but the underlying molecular mechanisms are largely unclear. Accumulating evidence points, however, to an important role of intracellular sphingosine-1-phosphate (S1P) metabolism in lipotoxicity-mediated disturbances of beta-cell function. In the present study, we compared the effects of an increased irreversible S1P degradation (S1P-lyase, SPL overexpression) with those associated with an enhanced S1P recycling (overexpression of S1P phosphatase 1, SGPP1) on LD formation and lipotoxicity in rat INS1E beta-cells. Interestingly, although both approaches led to a reduced S1P concentration, they had opposite effects on the susceptibility to FFA. Overexpression of SGPP1 prevented FFA-mediated caspase-3 activation by a mechanism involving an enhanced lipid storage capacity and prevention of oxidative stress. In contrast, SPL overexpression limited lipid droplet biogenesis, content and size, while accelerating lipophagy. This was associated with FFA-induced hydrogen peroxide formation, mitochondrial fragmentation and dysfunction, as well as ER stress. These changes coincided with upregulation of proapoptotic ceramides, but were independent of lipid peroxidation rate. Also in human EndoC-ßH1 beta-cells suppression of SPL with simultaneous overexpression of SGPP1 led to a similar and even more pronounced LD phenotype as that in INS1E-SGPP1 cells. Thus, intracellular S1P turnover significantly regulates LD content and size, and influences beta-cell sensitivity to FFA.

2.
Bioorg Med Chem Lett ; 94: 129464, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37634760

RESUMO

Simplified analogues of the myxobacterial polyketide ajudazol were obtained by synthesis and evaluated for their biological activities. Potent simplified 5-lipoxygenase inhibitors were identified. Moreover, strong antiproliferative and apoptotic activities were observed in brain cancer cell lines at low nano- to micromolar concentrations.


Assuntos
Neoplasias Encefálicas , Inibidores de Lipoxigenase , Neuroblastoma , Humanos , Araquidonato 5-Lipoxigenase , Linhagem Celular , Neuroblastoma/tratamento farmacológico , Inibidores de Lipoxigenase/química , Inibidores de Lipoxigenase/farmacologia
3.
Cells ; 12(14)2023 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-37508508

RESUMO

Astrocytes are critical players in brain health and disease. Brain pathologies and lesions are usually accompanied by astroglial alterations known as reactive astrogliosis. Sphingosine 1-phosphate lyase (SGPL1) catalysis, the final step in sphingolipid catabolism, irreversibly cleaves its substrate sphingosine 1-phosphate (S1P). We have shown that neural ablation of SGPL1 causes accumulation of S1P and hence neuronal damage, cognitive deficits, as well as microglial activation. Moreover, the S1P/S1P-receptor signaling axis enhances ATP production in SGPL1-deficient astrocytes. Using immunohistochemical methods as well as RNA Seq and CUT&Tag we show how S1P signaling causes activation of the astrocytic purinoreceptor P2Y1 (P2Y1R). With specific pharmacological agonists and antagonists, we uncover the P2Y1R as the key player in S1P-induced astrogliosis, and DDX3X mediated the activation of the NLRP3 inflammasome, including caspase-1 and henceforward generation of interleukin-1ß (IL-1ß) and of other proinflammatory cytokines. Our results provide a novel route connecting S1P metabolism and signaling with astrogliosis and the activation of the NLRP3 inflammasome, a central player in neuroinflammation, known to be crucial for the pathogenesis of numerous brain illnesses. Thus, our study opens the door for new therapeutic strategies surrounding S1P metabolism and signaling in the brain.


Assuntos
Inflamassomos , Liases , Encéfalo , Gliose , Proteína 3 que Contém Domínio de Pirina da Família NLR , Animais , Camundongos
4.
Pharmacol Ther ; 244: 108381, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36907249

RESUMO

Lipids are essential structural and functional components of the central nervous system (CNS). Sphingolipids are ubiquitous membrane components which were discovered in the brain in the late 19th century. In mammals, the brain contains the highest concentration of sphingolipids in the body. Sphingosine 1-phosphate (S1P) derived from membrane sphingolipids evokes multiple cellular responses which, depending on its concentration and localization, make S1P a double-edged sword in the brain. In the present review we highlight the role of S1P in brain development and focus on the often contrasting findings regarding its contributions to the initiation, progression and potential recovery of different brain pathologies, including neurodegeneration, multiple sclerosis (MS), brain cancers, and psychiatric illnesses. A detailed understanding of the critical implications of S1P in brain health and disease may open the door for new therapeutic options. Thus, targeting S1P-metabolizing enzymes and/or signaling pathways might help overcome, or at least ameliorate, several brain illnesses.


Assuntos
Cloridrato de Fingolimode , Esfingosina , Animais , Humanos , Esfingosina/metabolismo , Encéfalo/metabolismo , Lisofosfolipídeos/metabolismo , Esfingolipídeos , Receptores de Lisoesfingolipídeo/metabolismo , Mamíferos/metabolismo
5.
Int J Mol Sci ; 24(5)2023 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-36902011

RESUMO

Astrocytes are critical players in brain health and disease. Sphingosine-1-phosphate (S1P), a bioactive signaling lipid, is involved in several vital processes, including cellular proliferation, survival, and migration. It was shown to be crucial for brain development. Its absence is embryonically lethal, affecting, inter alia, the anterior neural tube closure. However, an excess of S1P due to mutations in S1P-lyase (SGPL1), the enzyme responsible for its constitutive removal, is also harmful. Of note, the gene SGPL1 maps to a region prone to mutations in several human cancers and also in S1P-lyase insufficiency syndrome (SPLIS) characterized by several symptoms, including peripheral and central neurological defects. Here, we investigated the impact of S1P on astrocytes in a mouse model with the neural-targeted ablation of SGPL1. We found that SGPL1 deficiency, and hence the accumulation of its substrate, S1P, causes the elevated expression of glycolytic enzymes and preferentially directs pyruvate into the tricarboxylic acid (TCA) cycle through its receptors (S1PR2,4). In addition, the activity of TCA regulatory enzymes was increased, and consequently, so was the cellular ATP content. The high energy load activates the mammalian target of rapamycin (mTOR), thus keeping astrocytic autophagy in check. Possible consequences for the viability of neurons are discussed.


Assuntos
Astrócitos , Esfingosina , Camundongos , Animais , Humanos , Astrócitos/metabolismo , Esfingosina/metabolismo , Encéfalo/metabolismo , Lisofosfolipídeos/metabolismo , Aldeído Liases/genética , Autofagia/fisiologia , Trifosfato de Adenosina/metabolismo , Mamíferos/metabolismo
6.
Mol Oncol ; 16(20): 3642-3653, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35973936

RESUMO

Sphingosine-1-phosphate (S1P), a bioactive signaling lipid, is involved in several vital processes, including cellular proliferation, survival and migration, as well as neovascularization and inflammation. Its critical role in the development and progression of cancer is well documented. The metabolism of S1P, which exerts its effect mainly via five G protein-coupled receptors (S1PR1-5 ), is tightly regulated. S1P-lyase (SGPL1) irreversibly cleaves S1P in the final step of sphingolipid catabolism and exhibits remarkably decreased enzymatic activity in tumor samples. In this study, we used SGPL1-deficient (Sgpl1-/- ) mouse embryonic fibroblasts (MEFs) and investigated the impact of S1P on glucose metabolism. Accumulated S1P activates, via its receptors (S1PR1-3 ), hypoxia-inducible factor 1 and stimulates the expression of proteins involved in glucose uptake and breakdown, indicating that Sgpl1-/- cells, like cancer cells, prefer to convert glucose to lactate even in the presence of oxygen. Accordingly, their rate of proliferation is significantly increased. Activation of the Akt/mTOR pathway and hence down-regulation of autophagy indicate that these changes do not negatively affect the cellular energy status. In summary, we report on a newly identified role of the S1P/S1PR1-3 axis in glucose metabolism in SGPL1-deficient MEFs.


Assuntos
Aldeído Liases , Fibroblastos , Proteínas Proto-Oncogênicas c-akt , Animais , Camundongos , Carcinogênese/metabolismo , Transformação Celular Neoplásica/metabolismo , Fibroblastos/metabolismo , Glucose/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , Lactatos/metabolismo , Lisofosfolipídeos/metabolismo , Oxigênio , Proteínas Proto-Oncogênicas c-akt/metabolismo , Esfingolipídeos/metabolismo , Esfingosina/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Aldeído Liases/genética , Camundongos Knockout
7.
EMBO Rep ; 23(5): e52606, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-35297148

RESUMO

Mitochondrial dysfunction can either extend or decrease Caenorhabditis elegans lifespan, depending on whether transcriptionally regulated responses can elicit durable stress adaptation to otherwise detrimental lesions. Here, we test the hypothesis that enhanced metabolic flexibility is sufficient to circumvent bioenergetic abnormalities associated with the phenotypic threshold effect, thereby transforming short-lived mitochondrial mutants into long-lived ones. We find that CEST-2.2, a carboxylesterase mainly localizes in the intestine, may stimulate the survival of mitochondrial deficient animals. We report that genetic manipulation of cest-2.2 expression has a minor lifespan impact on wild-type nematodes, whereas its overexpression markedly extends the lifespan of complex I-deficient gas-1(fc21) mutants. We profile the transcriptome and lipidome of cest-2.2 overexpressing animals and show that CEST-2.2 stimulates lipid metabolism and fatty acid beta-oxidation, thereby enhancing mitochondrial respiratory capacity through complex II and LET-721/ETFDH, despite the inherited genetic lesion of complex I. Together, our findings unveil a metabolic pathway that, through the tissue-specific mobilization of lipid deposits, may influence the longevity of mitochondrial mutant C. elegans.


Assuntos
Proteínas de Caenorhabditis elegans , Longevidade , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Metabolismo dos Lipídeos/genética , Longevidade/genética , Mitocôndrias/metabolismo
8.
Mol Oncol ; 15(12): 3256-3279, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34289244

RESUMO

Cancer development is a multistep process in which cells must overcome a series of obstacles before they can become fully developed tumors. First, cells must develop the ability to proliferate unchecked. Once this is accomplished, they must be able to invade the neighboring tissue, as well as provide themselves with oxygen and nutrients. Finally, they must acquire the ability to detach from the newly formed mass in order to spread to other tissues, all the while evading an immune system that is primed for their destruction. Furthermore, increased levels of inflammation have been shown to be linked to the development of cancer, with sites of chronic inflammation being a common component of tumorigenic microenvironments. In this Review, we give an overview of the impact of sphingolipid metabolism in cancers, from initiation to metastatic dissemination, as well as discussing immune responses and resistance to treatments. We explore how sphingolipids can either help or hinder the progression of cells from a healthy phenotype to a cancerous one.


Assuntos
Neoplasias , Esfingolipídeos , Ceramidas/metabolismo , Humanos , Lisofosfolipídeos/metabolismo , Neoplasias/patologia , Esfingolipídeos/metabolismo , Esfingosina/metabolismo , Microambiente Tumoral
9.
Anal Chem ; 93(8): 3867-3875, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33577289

RESUMO

Matrix-assisted laser desorption ionization-mass spectrometry imaging (MALDI-MSI) is an emerging label-free method for mapping the distribution of diverse molecular species in tissue sections. Despite recent progress in MALDI-MSI analyses of lipids, it is still difficult to visualize minor bioactive lipids including lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P). Here, we have developed a novel on-tissue derivatization method using Phos-tag, a zinc complex that specifically binds to a phosphate monoester group. MALDI-MSI with Phos-tag derivatization made it possible to image LPA and S1P in the murine brain. Furthermore, we were able to visualize other low-abundance lipids containing phosphate monoester, such as phosphatidic acid and ceramide-1-phosphate. Compared with conventional MALDI-MS, this derivatization produced LPA images with high spatial accuracy discriminating LPA artificially produced during MALDI-MS analysis. In mice with deficiencies in enzymes that degrade LPA and S1P, we observed marked S1P and/or LPA accumulation in specific regions of the brain. Thus, the present study provides a simple and optimal way to reveal the spatial localization of potent bioactive lipid phosphates such as LPA and S1P in tissues.


Assuntos
Lipídeos , Fosfatos , Animais , Camundongos , Piridinas , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
10.
Pharmacol Res ; 164: 105389, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33352226

RESUMO

The compatible solute ectoine is one of the most abundant and powerful cytoprotectant in the microbial world. Due to its unique ability to stabilize biological membranes and macromolecules it has been successfully commercialized as ingredient of various over-the-counter drugs, achieving primarily epithelial protection. While trying to elucidate the mechanism of its cell protective properties in in-vitro studies, a significant anti-inflammatory effect was documented for the small molecule. The tissue protective potential of ectoine considerably improved organ quality during preservation. In addition, ectoine and derivatives have been demonstrated to significantly decrease inflammatory cytokine production, thereby alleviating the inflammatory response following organ transplantation, and launching new therapeutic options for pathologies such as Inflammatory Bowel Disease (IBD) and Chronic Obstructive Pulmonary Disease (COPD). In this review, we aim to summarize the knowledge of this fairly nascent field of the anti-inflammatory potential of diverse ectoines. We also point out that this promising field faces challenges in its biochemical and molecular substantiations, including defining the molecular mechanisms of the observed effects and their regulation. However, based on their potent cytoprotective, anti-inflammatory, and non-toxic properties we believe that ectoines represent promising candidates for risk free interventions in inflammatory pathologies with steeply increasing demands for new therapeutics.


Assuntos
Diamino Aminoácidos/administração & dosagem , Anti-Inflamatórios/administração & dosagem , Doenças Inflamatórias Intestinais/tratamento farmacológico , Pneumopatias/tratamento farmacológico , Diamino Aminoácidos/biossíntese , Diamino Aminoácidos/farmacocinética , Animais , Anti-Inflamatórios/farmacocinética , Disponibilidade Biológica , Sistemas de Liberação de Medicamentos , Eucariotos/metabolismo , Humanos , Células Procarióticas/metabolismo
11.
Cells ; 9(11)2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33153026

RESUMO

It is an honor for us to dedicate this Special Issue to our dearest friend Lina Obeid, who was not only a pioneer in the field of sphingolipids, but also a remarkable personality [...].


Assuntos
Doença , Esfingolipídeos/uso terapêutico , Ensaios Clínicos como Assunto , Humanos , Transdução de Sinais
12.
Cells ; 9(10)2020 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-32998447

RESUMO

We have shown that sphingosine 1-phosphate (S1P) generated by sphingosine kinase 2 (SK2) is toxic in neurons lacking S1P-lyase (SGPL1), the enzyme that catalyzes its irreversible cleavage. Interestingly, patients harboring mutations in the gene encoding this enzyme (SGPL1) often present with neurological pathologies. Studies in a mouse model with a developmental neural-specific ablation of SGPL1 (SGPL1fl/fl/Nes) confirmed the importance of S1P metabolism for the presynaptic architecture and neuronal autophagy, known to be essential for brain health. We now investigated in SGPL1-deficient murine brains two other factors involved in neurodegenerative processes, namely tau phosphorylation and histone acetylation. In hippocampal and cortical slices SGPL1 deficiency and hence S1P accumulation are accompanied by hyperphosphorylation of tau and an elevated acetylation of histone3 (H3) and histone4 (H4). Calcium chelation with BAPTA-AM rescued both tau hyperphosphorylation and histone acetylation, designating calcium as an essential mediator of these (patho)physiological functions of S1P in the brain. Studies in primary cultured neurons and astrocytes derived from SGPL1fl/fl/Nes mice revealed hyperphosphorylated tau only in SGPL1-deficient neurons and increased histone acetylation only in SGPL1-deficient astrocytes. Both could be reversed to control values with BAPTA-AM, indicating the close interdependence of S1P metabolism, calcium homeostasis, and brain health.


Assuntos
Aldeído Liases/genética , Degeneração Neural/genética , Tauopatias/genética , Proteínas tau/genética , Acetilação , Animais , Autofagia/genética , Cálcio/metabolismo , Histonas/genética , Humanos , Lisofosfolipídeos/genética , Camundongos , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/metabolismo , Neurônios/patologia , Processamento de Proteína Pós-Traducional/genética , Esfingosina/análogos & derivados , Esfingosina/genética , Tauopatias/metabolismo , Tauopatias/patologia
13.
Protoplasma ; 257(6): 1709-1724, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32851422

RESUMO

Bird feather lipids are usually attributed to the oily secretion product of the uropygial (preen) gland. We have observed, however, that feathers exhibit a strong reaction with osmium tetroxide (OsO4), even after treatment with detergents. This leads us to postulate the existence of endogenous feather lipids distinct from preen gland lipids. In order to substantiate our hypothesis, we investigated down feathers from a 1-day-old chicken as their uropgygial gland is not functionally active. The results confirmed the osmiophilic reaction, which was concentrated in the center of barbs and strongly reduced after lipid extraction. In these lipid extracts, we identified using thin layer chromatography, cholesterol, various ceramides, glycolipids, phospholipids, and fatty acids, which closely resembled the lipid composition of the water barrier in the chicken-cornified epidermal envelope. This composition is clearly distinct from chicken uropygeal gland secretion (UGS) known to consist of fatty alcohols as part of aliphatic monoester waxes and of free, predominantly saturated, fatty acids. A filter assay showed a strong reactivity between OsO4 and the fatty acids C18:1 and C18:2 and with feather lipid extracts, but not with UGS. These observations were confirmed by gas chromatography detecting unsaturated fatty acids including C18:1 and C18:2 as well as cholesterol exclusively in chicken feathers. Our results indicate that (1) endogenous lipids are detectable in chicken feathers and distinct from UGS and (2) in analogy to the morphogenesis of the cornified envelope of chicken feather lipids that may have derived from cellular feather-precursors, apparently enduring the specific cell death during developmental feather cornification.


Assuntos
Plumas/química , Lipídeos/química , Glândulas Sebáceas/química , Animais , Galinhas
14.
Adv Drug Deliv Rev ; 159: 232-244, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32360155

RESUMO

Lipids play an important role in neurodegeneration, neuroinflammation, and psychiatric disorders and an imbalance in sphingolipid levels is associated with disease. Although early diagnosis and intervention of these disorders would clearly have favorable long-term outcomes, no diagnostic tests currently exist that can accurately identify people at risk. Reliable prognostic biomarkers that are easily accessible would be beneficial to determine therapy and treatment response in clinical trials. Recent advances in lipidomic investigation methods have greatly progressed the knowledge of sphingolipids in neurodegenerative and psychiatric disorders over the past decades although more longitudinal studies are needed to understand its exact role in these disorders to be used as potential tools in the clinic. In this review, we give an overview of the current knowledge of sphingolipids in neurodegenerative and psychiatric disorders and explore recent advances in investigation methods. Finally, the potential of sphingolipid metabolism products and signaling molecules as potential biomarkers for diagnosis, prognostic, or surrogate markers of treatment response is discussed.


Assuntos
Encefalite/metabolismo , Transtornos Mentais/metabolismo , Doenças Neurodegenerativas/metabolismo , Esfingolipídeos/metabolismo , Animais , Biomarcadores/metabolismo , Encefalite/diagnóstico , Encefalite/terapia , Humanos , Lipidômica , Transtornos Mentais/diagnóstico , Transtornos Mentais/terapia , Doenças Neurodegenerativas/diagnóstico , Doenças Neurodegenerativas/terapia , Prognóstico
15.
Artigo em Inglês | MEDLINE | ID: mdl-32283310

RESUMO

Sphingosine-1-phosphate (S1P) is not only a catabolic intermediate of all sphingolipids but also an evolutionary conserved bioactive lipid with critical functions in cell survival, differentiation, and migration as well as in immunity and angiogenesis. S1P-lyase (SGPL1) irreversibly cleaves S1P in the final step of sphingolipid catabolism. As sphingoid bases and their 1-phosphates are not only metabolic intermediates but also highly bioactive lipids that modulate a wide range of physiological processes, it would be predicted that their elevation might induce adjustments in other facets of sphingolipid metabolism and/or alter cell behavior. We actually found in a previous study that in terminally differentiated neurons SGPL1 deficiency increases sphingolipid formation via recycling at the expense of de novo synthesis. We now investigated whether and how SGPL1 deficiency affects the metabolism of (glyco)sphingolipids in mouse embryonic fibroblasts (MEFs). According to our previous experiments in neurons, we found a strong accumulation of S1P in SGPL1-deficient MEFs. Surprisingly, a completely different situation arose as we analyzed sphingolipid metabolism in this non-differentiated cell type. The production of biosynthetic precursors of complex glycosphingolipids including ceramide, glucosylceramide and also ganglioside GM3 via de novo synthesis and recycling pathway was substantially increased whereas the amount of more complex gangliosides dropped significantly.


Assuntos
Aldeído Liases/deficiência , Aldeído Liases/metabolismo , Carcinogênese , Gangliosídeos/biossíntese , Animais , Células Cultivadas , Gangliosídeos/análise , Camundongos , Camundongos Knockout
16.
Glia ; 67(10): 1859-1872, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31231866

RESUMO

Microglia mediated responses to neuronal damage in the form of neuroinflammation is a common thread propagating neuropathology. In this study, we investigated the microglial alterations occurring as a result of sphingosine 1-phosphate (S1P) accumulation in neural cells. We evidenced increased microglial activation in the brains of neural S1P-lyase (SGPL1) ablated mice (SGPL1fl/fl/Nes ) as shown by an activated and deramified morphology and increased activation markers on microglia. In addition, an increase of pro-inflammatory cytokines in sorted and primary cultured microglia generated from SGPL1 deficient mice was noticed. Further, we assessed autophagy, one of the major mechanisms in the brain that keeps inflammation in check. Indeed, microglial inflammation was accompanied by defective microglial autophagy in SGPL1 ablated mice. Rescuing autophagy by treatment with rapamycin was sufficient to decrease interleukin 6 (IL-6) but not tumor necrosis factor (TNF) secretion in cultured microglia. Rapamycin mediated decrease of IL-6 secretion suggests a particular mechanistic target of rapamycin (mTOR)-IL-6 link and appeared to be microglia specific. Using pharmacological inhibitors of the major receptors of S1P expressed in the microglia, we identified S1P receptor 2 (S1PR2) as the mediator of both impaired autophagy and proinflammatory effects. In line with these results, the addition of exogenous S1P to BV2 microglial cells showed similar effects as those observed in the genetic knock out of SGPL1 in the neural cells. In summary, we show a novel role of the S1P-S1PR2 axis in the microglia of mice with neural-targeted SGPL1 ablation and in BV2 microglial cell line exogenously treated with S1P.


Assuntos
Aldeído Liases/metabolismo , Autofagia/fisiologia , Inflamação/metabolismo , Microglia/metabolismo , Aldeído Liases/antagonistas & inibidores , Aldeído Liases/genética , Animais , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Inflamação/patologia , Interleucina-6/metabolismo , Camundongos Transgênicos , Microglia/patologia , Receptores de Esfingosina-1-Fosfato/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
17.
Biol Chem ; 399(8): 837-850, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-29908127

RESUMO

In mammals, the brain exhibits the highest lipid content in the body next to adipose tissue. Complex sphingolipids are characteristic compounds of neuronal membranes. Vital neural functions including information flux and transduction occur along these membranes. It is therefore not surprising that neuronal function and survival is dependent on the metabolism of these lipids. Autophagy is a critical factor for the survival of post-mitotic neurons. On the one hand, it fulfils homeostatic and waste-recycling functions and on the other hand, it constitutes an effective strategy to eliminate harmful proteins that cause neuronal death. A growing number of experimental data indicate that several sphingolipids as well as enzymes catalyzing their metabolic transformations efficiently but very differently affect neuronal autophagy and hence survival. This review attempts to elucidate the roles and mechanisms of sphingolipid metabolism with regard to the regulation of autophagy and its consequences for brain physiology and pathology.


Assuntos
Autofagia , Encéfalo/citologia , Encéfalo/metabolismo , Esfingolipídeos/metabolismo , Animais , Encéfalo/patologia , Humanos , Neurônios/citologia , Neurônios/metabolismo , Neurônios/patologia
18.
Autophagy ; 13(5): 885-899, 2017 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-28521611

RESUMO

Macroautophagy/autophagy defects have been identified as critical factors underlying the pathogenesis of neurodegenerative diseases. The roles of the bioactive signaling lipid sphingosine-1-phosphate (S1P) and its catabolic enzyme SGPL1/SPL (sphingosine phosphate lyase 1) in autophagy are increasingly recognized. Here we provide in vitro and in vivo evidence for a previously unidentified route through which SGPL1 modulates autophagy in neurons. SGPL1 cleaves S1P into ethanolamine phosphate, which is directed toward the synthesis of phosphatidylethanolamine (PE) that anchors LC3-I to phagophore membranes in the form of LC3-II. In the brains of SGPL1fl/fl/Nes mice with developmental neural specific SGPL1 ablation, we observed significantly reduced PE levels. Accordingly, alterations in basal and stimulated autophagy involving decreased conversion of LC3-I to LC3-II and increased BECN1/Beclin-1 and SQSTM1/p62 levels were apparent. Alterations were also noticed in downstream events of the autophagic-lysosomal pathway such as increased levels of lysosomal markers and aggregate-prone proteins such as APP (amyloid ß [A4] precursor protein) and SNCA/α-synuclein. In vivo profound deficits in cognitive skills were observed. Genetic and pharmacological inhibition of SGPL1 in cultured neurons promoted these alterations, whereas addition of PE was sufficient to restore LC3-I to LC3-II conversion, and control levels of SQSTM1, APP and SNCA. Electron and immunofluorescence microscopy showed accumulation of unclosed phagophore-like structures, reduction of autolysosomes and altered distribution of LC3 in SGPL1fl/fl/Nes brains. Experiments using EGFP-mRFP-LC3 provided further support for blockage of the autophagic flux at initiation stages upon SGPL1 deficiency due to PE paucity. These results emphasize a formerly overlooked direct role of SGPL1 in neuronal autophagy and assume significance in the context that autophagy modulators hold an enormous therapeutic potential in the treatment of neurodegenerative diseases.


Assuntos
Aldeído Liases/metabolismo , Peptídeos beta-Amiloides/metabolismo , Autofagia/fisiologia , Neurônios/metabolismo , Fosfatidiletanolaminas/metabolismo , Animais , Encéfalo/metabolismo , Lisofosfolipídeos/metabolismo , Lisossomos/metabolismo , Camundongos , Esfingosina/análogos & derivados , Esfingosina/metabolismo
19.
Sci Rep ; 7: 43575, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28262793

RESUMO

Sphingosine-1-phosphate (S1P) lyase irreversibly cleaves S1P, thereby catalysing the ultimate step of sphingolipid degradation. We show here that embryonic fibroblasts from S1P lyase-deficient mice (Sgpl1-/--MEFs), in which S1P and sphingosine accumulate, have features of Niemann-Pick disease type C (NPC) cells. In the presence of serum, overall cholesterol content was elevated in Sgpl1-/--MEFs, due to upregulation of the LDL receptor and enhanced cholesterol uptake. Despite this, activation of sterol regulatory element-binding protein-2 was increased in Sgpl1-/--MEFs, indicating a local lack of cholesterol at the ER. Indeed, free cholesterol was retained in NPC1-containing vesicles, which is a hallmark of NPC. Furthermore, upregulation of amyloid precursor protein in Sgpl1-/--MEFs was mimicked by an NPC1 inhibitor in Sgpl1+/+-MEFs and reduced by overexpression of NPC1. Lysosomal pH was not altered by S1P lyase deficiency, similar to NPC. Interestingly, lysosomal Ca2+ content and bafilomycin A1-induced [Ca2+]i increases were enhanced in Sgpl1-/--MEFs, contrary to NPC. These results show that both a primary defect in cholesterol trafficking and S1P lyase deficiency cause overlapping phenotypic alterations, and challenge the present view on the role of sphingosine in lysosomal Ca2+ homeostasis.


Assuntos
Aldeído Liases/deficiência , Cálcio/metabolismo , Colesterol/metabolismo , Fibroblastos/metabolismo , Lisossomos/metabolismo , Doença de Niemann-Pick Tipo C/genética , Doença de Niemann-Pick Tipo C/metabolismo , Aldeído Liases/sangue , Animais , Biomarcadores , Modelos Animais de Doenças , Histona Desacetilases , Homeostase , Concentração de Íons de Hidrogênio , Espaço Intracelular/metabolismo , Camundongos , Camundongos Knockout , Doença de Niemann-Pick Tipo C/diagnóstico , Fenótipo
20.
Biochim Biophys Acta Biomembr ; 1859(9 Pt B): 1573-1582, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28315304

RESUMO

The physiological functions of sphingosine 1-phosphate (S1P) and its pathological roles in various diseases are increasingly being elucidated. Particularly, a growing body of literature has implicated S1P in the pathogenesis of brain related disorders. With the deciphering of more intricate aspects of S1P signalling, there is also a need to reconsider the notion of S1P only as a determinant of cell survival and proliferation. Further the concept of 'S1P-ceramide' balance as the controlling switch of cellular fate and functions needs to be refined. In this review, we focus on the brain related functions of S1P with special focus on its role in synaptic transmission, neuronal autophagy and neuroinflammation. The review also attempts to bring out the multi-faceted nature of S1P signalling aspects that makes it a 'double edged sword'. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.


Assuntos
Encefalopatias/etiologia , Lisofosfolipídeos/fisiologia , Esfingosina/análogos & derivados , Animais , Astrócitos/fisiologia , Autofagia , Humanos , Microglia/fisiologia , Oligodendroglia/fisiologia , Transdução de Sinais/fisiologia , Esfingosina/fisiologia , Transmissão Sináptica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA