Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 7(5): e37218, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22606352

RESUMO

Sphingosine-1-phosphate (S1P) activates a widely expressed family of G protein-coupled receptors, serves as a muscle trophic factor and activates muscle stem cells called satellite cells (SCs) through unknown mechanisms. Here we show that muscle injury induces dynamic changes in S1P signaling and metabolism in vivo. These changes include early and profound induction of the gene encoding the S1P biosynthetic enzyme SphK1, followed by induction of the catabolic enzyme sphingosine phosphate lyase (SPL) 3 days later. These changes correlate with a transient increase in circulating S1P levels after muscle injury. We show a specific requirement for SphK1 to support efficient muscle regeneration and SC proliferation and differentiation. Mdx mice, which serve as a model for muscular dystrophy (MD), were found to be S1P-deficient and exhibited muscle SPL upregulation, suggesting that S1P catabolism is enhanced in dystrophic muscle. Pharmacological SPL inhibition increased muscle S1P levels, improved mdx muscle regeneration and enhanced SC proliferation via S1P receptor 2 (S1PR2)-dependent inhibition of Rac1, thereby activating Signal Transducer and Activator of Transcription 3 (STAT3), a central player in inflammatory signaling. STAT3 activation resulted in p21 and p27 downregulation in a S1PR2-dependent fashion in myoblasts. Our findings suggest that S1P promotes SC progression through the cell cycle by repression of cell cycle inhibitors via S1PR2/STAT3-dependent signaling and that SPL inhibition may provide a therapeutic strategy for MD.


Assuntos
Lisofosfolipídeos/farmacologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Fator de Transcrição STAT3/metabolismo , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Células Satélites de Músculo Esquelético/metabolismo , Esfingosina/análogos & derivados , Animais , Proliferação de Células , Feminino , Lisofosfolipídeos/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Regeneração/efeitos dos fármacos , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/patologia , Transdução de Sinais/efeitos dos fármacos , Esfingosina/deficiência , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato
2.
Carcinogenesis ; 33(9): 1726-35, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22581840

RESUMO

Sphingadienes (SDs) derived from soy and other natural sphingolipids are cytotoxic to colon cancer cells via an Akt-dependent mechanism and reduce adenoma formation in Apc(Min/+) mice. Wnt signaling is fundamental to colon carcinogenesis and is the basis for spontaneous tumorigenesis in Apc(Min/+) mice and patients with familial adenomatous polyposis. In the present study, we investigated the impact of SDs on Wnt signaling. Oral SD administration reduced levels of active ß-catenin and Wnt targets c-Myc and cyclin D1 in Apc(Min/+) mouse intestinal tissues. Colon cancer cells treated with SDs exhibited reduced Wnt transcriptional activity, as well as reduced nuclear ß-catenin localization and subsequent reduction in active-ß-catenin levels. Further, we observed a decrease in phosphorylated (inactive) GSK3ß in SD-treated mice and colon cancer cells. Expression of constitutively active myristoylated-Akt or inactivation of GSK3ß using LiCl attenuated SD-mediated inhibition of Wnt transcriptional activity and active-ß-catenin levels. SDs exhibited additive effects with inhibitors of the phosphatidylinositol-3-kinase/Akt/mTOR pathway to induce cytotoxicity. Further, a combination regime of SDs and low-dose rapamycin decreased visible polyps in Apc(Min/+) mice and reduced the levels of Wnt target gene expression and mTOR target activation. SD-mediated inhibition of Akt and Wnt pathways and cytotoxicity in colon cancer cells was dependent upon the activity of protein phosphatase 2A, as shown by reversal of these effects by pretreatment with okadaic acid or calyculin A. Our cumulative findings indicate that SDs inhibit Wnt signaling through a protein phosphatase 2A/Akt/GSK3ß-dependent mechanism that may contribute to their chemopreventive effects in intestinal tumorigenesis.


Assuntos
Anticarcinógenos/farmacologia , Neoplasias do Colo/prevenção & controle , Quinase 3 da Glicogênio Sintase/fisiologia , Proteína Fosfatase 2/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Esfingolipídeos/farmacologia , Proteína Supressora de Tumor p53/fisiologia , Via de Sinalização Wnt/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/fisiologia , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Regulação para Baixo , Glicogênio Sintase Quinase 3 beta , Humanos , Elementos de Resposta , Transdução de Sinais , Via de Sinalização Wnt/fisiologia
3.
PLoS One ; 6(11): e26993, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22069480

RESUMO

Wnt proteins comprise a large family of secreted ligands implicated in a wide variety of biological roles. WntD has previously been shown to inhibit the nuclear accumulation of Dorsal/NF-κB protein during embryonic dorsal/ventral patterning and the adult innate immune response, independent of the well-studied Armadillo/ß-catenin pathway. In this paper, we present a novel phenotype for WntD mutant embryos, suggesting that this gene is involved in migration of primordial germ cells (PGC) to the embryonic gonad. Additionally, we describe a genetic suppressor/enhancer screen aimed at identifying genes required for WntD signal transduction, based on the previous observation that maternal overexpression of WntD results in lethally dorsalized embryos. Using an algorithm to narrow down our hits from the screen, we found two novel WntD signaling components: Fz4, a member of the Frizzled family, and the Drosophila Ceramide Kinase homolog, Dcerk. We show here that Dcerk and Dmulk (Drosophila Multi-substrate lipid kinase) redundantly mediate PGC migration. Our data are consistent with a model in which the activity of lipid phosphate phosphatases shapes a concentration gradient of ceramide-1-phosphate (C1P), the product of Dcerk, allowing proper PGC migration.


Assuntos
Movimento Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Testes Genéticos , Células Germinativas/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptores Acoplados a Proteínas G/genética , Animais , Animais Geneticamente Modificados , Southern Blotting , Western Blotting , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Ceramidas/metabolismo , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Elementos Facilitadores Genéticos , Feminino , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/genética , Metabolismo dos Lipídeos , Masculino , Filogenia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores Acoplados a Proteínas G/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Supressão Genética , beta Catenina/metabolismo
4.
Am J Physiol Heart Circ Physiol ; 300(5): H1753-61, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21335477

RESUMO

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that promotes cardiomyocyte survival and contributes to ischemic preconditioning. S1P lyase (SPL) is a stress-activated enzyme responsible for irreversible S1P catabolism. We hypothesized that SPL contributes to oxidative stress by depleting S1P pools available for cardioprotective signaling. Accordingly, we evaluated SPL inhibition as a strategy for reducing cardiac ischemia-reperfusion (I/R) injury. We measured SPL expression and enzyme activity in murine hearts. Basal SPL activity was low in wild-type cardiac tissue but was activated in response to 50 min of ischemia (n = 5, P < 0.01). Hearts of heterozygous SPL knockout mice exhibited reduced SPL activity, elevated S1P levels, smaller infarct size, and increased functional recovery after I/R compared with littermate controls (n = 5, P < 0.01). The small molecule tetrahydroxybutylimidazole (THI) is a Federal Drug Administration-approved food additive that inhibits SPL. When given overnight at 25 mg/l in drinking water, THI raised S1P levels and reduced SPL activity (n = 5, P < 0.01). THI reduced infarct size and enhanced hemodynamic recovery in response to 50 min of ischemia and to 40 min of reperfusion in ex vivo hearts (n = 7, P < .01). These data correlated with an increase in MAP kinase-interacting serine/threonine kinase 1, eukaryotic translation initiation factor 4E, and ribosomal protein S6 phosphorylation levels after I/R, suggesting that SPL inhibition enhances protein translation. Pretreatment with an S1P1 and S1P3 receptor antagonist partially reversed the effects of THI. These results reveal, for the first time, that SPL is an ischemia-induced enzyme that can be targeted as a novel strategy for preventing cardiac I/R injury.


Assuntos
Aldeído Liases/antagonistas & inibidores , Aldeído Liases/fisiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Aldeído Liases/genética , Animais , Inibidores Enzimáticos/uso terapêutico , Lisofosfolipídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Mutação/genética , Miocárdio/metabolismo , Estresse Oxidativo/fisiologia , Esfingosina/análogos & derivados , Esfingosina/metabolismo
5.
Nat Chem Biol ; 6(7): 489-97, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20559316

RESUMO

Sphingolipids comprise a complex family of naturally occurring molecules that are enriched in lipid rafts and contribute to their unique biochemical properties. Membrane sphingolipids also serve as a reservoir for bioactive metabolites including sphingosine, ceramide, sphingosine-1-phosphate and ceramide-1-phosphate. Among these, sphingosine-1-phosphate has emerged as a central regulator of mammalian biology. Sphingosine-1-phosphate is essential for mammalian brain and cardiac development and for maturation of the systemic circulatory system and lymphatics. In addition, sphingosine-1-phosphate contributes to trafficking and effector functions of lymphocytes and other hematopoietic cells and protects against various forms of tissue injury. However, sphingosine-1-phosphate is also an oncogenic lipid that promotes tumor growth and progression. Recent preclinical and clinical investigations using pharmacological agents that target sphingosine-1-phosphate, its receptors and the enzymes required for its biosynthesis and degradation demonstrate the promise and potential risks of modulating sphingosine-1-phosphate signaling in treatment strategies for autoimmunity, cancer, cardiovascular disease and other pathological conditions.


Assuntos
Lisofosfolipídeos/metabolismo , Esfingolipídeos/metabolismo , Esfingosina/análogos & derivados , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Lisofosfolipídeos/genética , Esfingolipídeos/genética , Esfingosina/genética , Esfingosina/metabolismo
6.
Cancer Res ; 69(24): 9457-64, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19934323

RESUMO

Sphingolipid metabolites regulate cell proliferation, migration, and stress responses. Alterations in sphingolipid metabolism have been proposed to contribute to carcinogenesis, cancer progression, and drug resistance. We identified a family of natural sphingolipids called sphingadienes and investigated their effects in colon cancer. We find that sphingadienes induce colon cancer cell death in vitro and prevent intestinal tumorigenesis in vivo. Sphingadienes exert their influence by blocking Akt translocation from the cytosol to the membrane, thereby inhibiting protein translation and promoting apoptosis and autophagy. Sphingadienes are orally available, are slowly metabolized through the sphingolipid degradative pathway, and show limited short-term toxicity. Thus, sphingadienes represent a new class of therapeutic and/or chemopreventive agents that blocks Akt signaling in neoplastic and preneoplastic cells.


Assuntos
Alcadienos/farmacologia , Neoplasias do Colo/prevenção & controle , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Esfingolipídeos/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Ativação Enzimática/efeitos dos fármacos , Células HCT116 , Células HT29 , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
Circ Res ; 103(10): 1164-72, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18849324

RESUMO

The lipid mediator sphingosine-1-phosphate (S1P), the product of sphingosine kinase (SPHK)-induced phosphorylation of sphingosine, is known to stabilize interendothelial junctions and prevent microvessel leakiness. Here, we investigated the role of SPHK1 activation in regulating the increase in pulmonary microvessel permeability induced by challenge of mice with lipopolysaccharide or thrombin ligation of protease-activating receptor (PAR)-1. Both lipopolysaccharide and thrombin increased mouse lung microvascular permeability and resulted in a delayed activation of SPHK1 that was coupled to the onset of restoration of permeability. In contrast to wild-type mice, Sphk1(-/-) mice showed markedly enhanced pulmonary edema formation in response to lipopolysaccharide and PAR-1 activation. Using endothelial cells challenged with thrombin concentration (50 nmol/L) that elicited a transient but reversible increase in endothelial permeability, we observed that increased SPHK1 activity and decreased intracellular S1P concentration preceded the onset of barrier recovery. Thus, we tested the hypothesis that released S1P in a paracrine manner activates its receptor S1P1 to restore the endothelial barrier. Knockdown of SPHK1 decreased basal S1P production and Rac1 activity but increased basal endothelial permeability. In SPHK1-depleted cells, PAR-1 activation failed to induce Rac1 activation but augmented RhoA activation and endothelial hyperpermeability response. Knockdown of S1P1 receptor in endothelial cells also enhanced the increase in endothelial permeability following PAR-1 activation. S1P treatment of Sphk1(-/-) lungs or SPHK1-deficient endothelial cells restored endothelial barrier function. Our results suggest the crucial role of activation of the SPHK1-->S1P-->S1P1 signaling pathway in response to inflammatory mediators in endothelial cells in regulating endothelial barrier homeostasis.


Assuntos
Permeabilidade Capilar , Mediadores da Inflamação/metabolismo , Junções Intercelulares/metabolismo , Pulmão/metabolismo , Lisofosfolipídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/genética , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Hemostáticos/efeitos adversos , Hemostáticos/farmacologia , Humanos , Mediadores da Inflamação/farmacologia , Junções Intercelulares/genética , Lipopolissacarídeos/toxicidade , Lisofosfolipídeos/genética , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Knockout , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/genética , Edema Pulmonar/metabolismo , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esfingosina/genética , Esfingosina/metabolismo , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato , Trombina/efeitos adversos , Trombina/farmacologia , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
8.
Biochim Biophys Acta ; 1781(9): 448-58, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18558101

RESUMO

Sphingosine-1-phosphate lyase (SPL) is a highly conserved enzyme that catalyses the final step of sphingolipid degradation, namely the irreversible cleavage of the carbon chain at positions 2-3 of a long-chain base phosphate (LCBP), thereby yielding a long-chain aldehyde and phosphoethanolamine. LCBPs are potent signaling molecules involved in cell proliferation, survival, migration, cell-cell interactions and cell stress responses. Therefore, tight regulation of LCBP signaling is required for proper cell function, and perturbations of this system can lead to alterations in biological processes including development, reproduction and physiology. SPL is a key enzyme in regulating the intracellular and circulating levels of LCBPs and is, therefore, gaining attention as a putative target for pharmacological intervention. This review provides an overview of our current understanding of SPL structure and function, mechanisms involved in SPL regulation and the role of SPL in development and disease.


Assuntos
Aldeído Liases/metabolismo , Doença , Esfingosina/metabolismo , Aldeído Liases/química , Aldeído Liases/genética , Animais , Apoptose , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Esfingosina/química
9.
Proc Natl Acad Sci U S A ; 105(1): 88-93, 2008 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18156367

RESUMO

The mammalian RBC lacks de novo lipid synthesis but maintains its membrane composition by rapid turnover of acyl moieties at the sn-2 position of phospholipids. Plasma-derived fatty acids are esterified to acyl-CoA by acyl-CoA synthetases and transferred to lysophospholipids by acyl-CoA:lysophospholipid acyltransferases. We report the characterization of three lysophosphatidylcholine (lysoPC) acyltransferases (LPCATs), products of the AYTL1, -2, and -3 genes. These proteins are three members of a LPCAT family, of which all three genes are expressed in an erythroleukemic cell line. Aytl2 mRNA was detected in mouse reticulocytes, and the presence of the product of the human ortholog was confirmed in adult human RBCs. The three murine Aytl proteins generated phosphatidylcholine from long-chain acyl-CoA and lysoPC when expressed in Escherichia coli membranes. Spliced variants of Aytl1, affecting a conserved catalytic motif, were identified. Calcium and magnesium modulated LPCAT activity of both Aytl1 and -2 proteins that exhibit EF-hand motifs at the C terminus. Characterization of the product of the Aytl2 gene as the phosphatidylcholine reacylating enzyme in RBCs represents the identification of a plasma membrane lysophospholipid acyltransferase and establishes the function of a LPCAT protein.


Assuntos
1-Acilglicerofosfocolina O-Aciltransferase/fisiologia , 1-Acilglicerofosfocolina O-Aciltransferase/genética , Sequência de Aminoácidos , Animais , Domínio Catalítico , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Clonagem Molecular , Eritrócitos/metabolismo , Escherichia coli/metabolismo , Ácidos Graxos/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Fosfatidilcolinas/química , Reticulócitos/metabolismo , Homologia de Sequência de Aminoácidos
10.
J Lipid Res ; 49(3): 597-606, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18156591

RESUMO

Sphingolipids comprise a complex group of lipids concentrated in membrane rafts and whose metabolites function as signaling molecules. Sphingolipids are conserved in Drosophila, in which their tight regulation is required for proper development and tissue integrity. In this study, we identified a new family of Drosophila sphingolipids containing two double bonds in the long chain base (LCB). The lipids were found at low levels in wild-type flies and accumulated markedly in Drosophila Sply mutants, which do not express sphingosine-1-phosphate lyase and are defective in sphingolipid catabolism. To determine the identity of the unknown lipids, purified whole fly lipid extracts were separated on a C18-HPLC column and analyzed using electrospray mass spectrometry. The lipids contain a LCB of either 14 or 16 carbons with conjugated double bonds at C4,6. The Delta(4,6)-sphingadienes were found as free LCBs, as phosphorylated LCBs, and as the sphingoid base in ceramides. The temporal and spatial accumulation of Delta(4,6)-sphingadienes in Sply mutants suggests that these lipids may contribute to the muscle degeneration observed in these flies.


Assuntos
Drosophila/química , Espectrometria de Massas por Ionização por Electrospray/métodos , Esfingolipídeos/análise , Aldeído Liases/deficiência , Aldeído Liases/metabolismo , Alcadienos , Animais , Mutação , Esfingolipídeos/química , Esfingolipídeos/metabolismo
11.
J Lipid Res ; 48(12): 2769-78, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17872591

RESUMO

Sphingosine-1-phosphate (S1P) lyase (SPL) catalyzes the conversion of S1P to ethanolamine phosphate and hexadecenal. This enzyme plays diverse roles in physiology and disease and, thus, may be useful as a disease marker and/or drug target. Unfortunately, the radioisotope-based assay currently used to quantify SPL activity is suboptimal. We have devised an assay using a commercially available omega(7-nitro-2-1,3-benzoxadiazol-4-yl)-d-erythro (NBD)-labeled fluorescent substrate. Alternatively, we provide a method for synthesis of the substrate from NBD-sphingosine. Enzyme activity is determined by following the formation of NBD-aldehyde product, which is isolated from unreacted substrate by lipid extraction and quantified after separation by HPLC using a C18 column. A fluorescent NBD-C18-sphingosine internal standard is used to control for extraction efficiency. The reaction is linear over 20 min and total protein concentrations of 20-200 mg/l. The sensitivity of the fluorescence assay is comparable to or better than that of the radioactive assay, and SPL levels as low as 8 pmol/mg/min were readily detected. Semicarbazide, a nonspecific SPL inhibitor, reduced SPL activity in vitro by approximately 70% using both standard and fluorescence methods. Product inhibition was not observed using ethanolamine phosphate and a commercially available source of hexadecenal. This method is suitable for quantifying SPL activity in a variety of cell and tissue sources.


Assuntos
Aldeído Liases/análise , Espectrometria de Fluorescência/métodos , Células 3T3-L1 , Aldeído Liases/química , Aldeído Liases/metabolismo , Animais , Catecóis/química , Linhagem Celular Tumoral , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Camundongos , Compostos Organometálicos/química
12.
Dev Biol ; 309(2): 329-41, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17706961

RESUMO

Sphingolipid signaling is thought to regulate apoptosis via mechanisms that are dependent on the concentration of ceramide relative to that of sphingosine-1-phosphate (S1P). This study reports defects in reproductive structures and function that are associated with enhanced apoptosis in Drosophila Sply05091 mutants that lack functional S1P lyase and thereby accumulate sphingolipid long chain base metabolites. Analyses of reproductive structures in these adult mutants unmasked multiple abnormalities, including supernumerary spermathecae, degenerative ovaries, and severely reduced testes. TUNEL assessment revealed increased cell death in mutant egg chambers at most oogenic stages and in affected mutant testes. These reproductive abnormalities and elevated gonadal apoptosis were also observed, to varying degrees, in other mutants affecting sphingolipid metabolism. Importantly, the reproductive defects seen in the Sply05091 mutants were ameliorated both by a second site mutation in the lace gene that restores long chain base levels towards normal and by genetic disruption of the proapoptotic genes reaper, hid and grim. These data thus provide the first evidence in Drosophila that accumulated sphingolipids trigger elevated levels of apoptosis via the modulation of known signaling pathways.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/enzimologia , Lisofosfolipídeos/metabolismo , Esfingolipídeos/metabolismo , Esfingosina/análogos & derivados , Aldeído Liases/genética , Aldeído Liases/metabolismo , Animais , Apoptose , Drosophila/genética , Proteínas de Drosophila/genética , Feminino , Genitália/anormalidades , Genitália/enzimologia , Larva , Masculino , Mutação , Esfingosina/metabolismo
13.
Mol Cell Biol ; 26(19): 7211-23, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16980623

RESUMO

Sphingosine kinase (Sphk) enzymes are important in intracellular sphingolipid metabolism as well as in the biosynthesis of sphingosine 1-phosphate (S1P), an extracellular lipid mediator. Here, we show that Sphk1 is expressed and is required for small intestinal tumor cell proliferation in Apc Min/+ mice. Adenoma size but not incidence was dramatically reduced in Apc Min/+ Sphk(-/-) mice. Concomitantly, epithelial cell proliferation in the polyps was significantly attenuated, suggesting that Sphk1 regulates adenoma progression. Although the S1P receptors (S1P1R, S1P2R, and S1P3R) are expressed, polyp incidence or size was unaltered in Apc Min/+ S1p2r(-/-), Apc Min/+ S1p3r(-/-), and Apc Min/+ S1p1r(+/-) bigenic mice. These data suggest that extracellular S1P signaling via its receptors is not involved in adenoma cell proliferation. Interestingly, tissue sphingosine content was elevated in the adenomas of Apc Min/+ Sphk1(-/-) mice, whereas S1P levels were not significantly altered. Concomitantly, epithelial cell proliferation and the expression of the G1/S cell cycle regulator CDK4 and c-myc were diminished in the polyps of Apc Min/+ Sphk1(-/-) mice. In rat intestinal epithelial (RIE) cells in vitro, Sphk1 overexpression enhanced cell cycle traverse at the G1/S boundary. In addition, RIE cells treated with sphingosine but not C6-ceramide exhibited reduced cell proliferation, reduced retinoblastoma protein phosphorylation, and cyclin-dependent kinase 4 (Cdk4) expression. Our findings suggest that Sphk1 plays a critical role in intestinal tumor cell proliferation and that inhibitors of Sphk1 may be useful in the control of intestinal cancer.


Assuntos
Pólipos Adenomatosos/enzimologia , Pólipos Adenomatosos/patologia , Neoplasias Gastrointestinais/enzimologia , Neoplasias Gastrointestinais/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Pólipos Adenomatosos/genética , Animais , Apoptose/fisiologia , Proteínas de Ciclo Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células , Regulação para Baixo/genética , Fase G1/fisiologia , Neoplasias Gastrointestinais/genética , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/patologia , Lisofosfolipídeos/metabolismo , Camundongos , Neovascularização Patológica , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores de Lisoesfingolipídeo/genética , Fase S/fisiologia , Esfingosina/análogos & derivados , Esfingosina/metabolismo
14.
J Biol Chem ; 280(18): 18403-10, 2005 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-15734735

RESUMO

Sphingosine-1-phosphate is a bioactive sphingolipid that regulates proliferation, differentiation, migration, and apoptosis. Sphingosine-1-phosphate is irreversibly degraded by the highly conserved enzyme sphingosine-1-phosphate lyase. Recent studies have suggested that sphingosine-1-phosphate lyase expression affects animal development and cell fate decisions. Despite its crucial role, mechanisms affecting expression of sphingosine-1-phosphate lyase remain poorly understood. In this study, regulation of sphingosine-1-phosphate lyase gene expression was investigated in Caenorhabditis elegans, where lyase expression is spatially restricted to cells of the developing and adult gut and is essential for normal development. Deletion analysis and generation of transgenic worms combined with fluorescence microscopy identified a 350-nucleotide sequence upstream of the ATG start site necessary for maximal lyase expression in adult worms. Site-specific mutagenesis of a GATA transcription factor-binding motif in the promoter led to loss of reporter expression. Knockdown of the gut-specific GATA transcription factor ELT-2 by RNA interference similarly led to loss of reporter expression. ELT-2 interacted with the GATA factor-binding motif in vitro and was also capable of driving expression of a Caenorhabditis elegans lyase promoter-beta-galactosidase reporter in a heterologous yeast system. These studies demonstrate that ELT-2 regulates sphingosine-1-phosphate lyase expression in vivo. Additionally, we demonstrate that the human sphingosine-1-phosphate lyase gene is regulated by a GATA transcription factor. Overexpression of GATA-4 led to both an increase in activity of a reporter gene as well as an increase in endogenous sphingosine-1-phosphate lyase protein.


Assuntos
Aldeído Liases/biossíntese , Aldeído Liases/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Aldeído Liases/metabolismo , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/biossíntese , Proteínas de Caenorhabditis elegans/genética , Linhagem Celular , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Fatores de Transcrição GATA , Fator de Transcrição GATA4 , Humanos
15.
Proc Natl Acad Sci U S A ; 101(51): 17825-30, 2004 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-15596715

RESUMO

gamma-Tocopherol (gammaT), the predominant form of vitamin E in diets, but not alpha-tocopherol, the major vitamin E form in tissues and supplements, inhibits proliferation of prostate cancer cells (LNCaP and PC-3) and lung cancer cells (A549). In contrast, at similar concentrations, gammaT has no effect on normal prostate epithelial cells. Combinations of some vitamin E forms, such as gammaT and delta-tocopherol, exhibit additive or synergistic inhibitory effects. In this study, gammaT or its combination with delta-tocopherol induced apoptosis in androgen-sensitive prostate LNCaP, but not in androgen-resistant PC-3 cells, by the induction of cytochrome c release, activation of caspase 9 and caspase 3, cleavage of poly-ADP-ribose polymerase (PARP), and involvement of caspase-independent pathways. Myriocin and fumonisin B1, specific inhibitors of key enzymes (serine palmitoyltransferase and dihydroceramide synthase, respectively) in de novo synthesis of sphingolipids, significantly protected cells from gammaT-induced DNA fragmentation, cytochrome c release, PARP cleavage, and the formation of active caspase 3. Compared with vehicle-treated controls, gammaT treatment led to pronounced dihydroceramide and dihydrosphingosine accumulation, which preceded morphological and biochemical manifestations of apoptosis. In contrast, ceramide and shpingosine levels did not increase until day 3, when substantial cell death took place. Our study demonstrates that gammaT and mixed vitamin E forms induce cell death by interrupting the de novo sphingolipid pathway in a prostate cancer cell line. Thus, certain vitamin E forms may be valuable as anticancer agents.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Esfingolipídeos/biossíntese , gama-Tocoferol/farmacologia , Ácido Araquidônico/farmacologia , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Células Epiteliais/citologia , Ácidos Graxos Monoinsaturados/farmacologia , Humanos , Ácido Linoleico/farmacologia , Masculino , Estrutura Molecular , Próstata/citologia , Próstata/efeitos dos fármacos , Esfingolipídeos/metabolismo
16.
J Biol Chem ; 279(13): 12685-94, 2004 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-14722126

RESUMO

Sphingosine kinase is a highly conserved enzyme that catalyzes the synthesis of sphingosine 1-phosphate and reduces cellular levels of sphingosine and ceramide. Although ceramide is pro-apoptotic and sphingosine is generally growth-inhibitory, sphingosine 1-phosphate signaling promotes cell proliferation, survival, and migration. Sphingosine kinase is thus in a strategic position to regulate important cell fate decisions which may contribute to normal animal development. To facilitate studies examining the potential role of sphingosine kinase and long chain base metabolism in Drosophila development, we characterized two putative Drosophila sphingosine kinase genes, Sk1 and Sk2. Both genes functionally and biochemically complement a yeast sphingosine kinase mutant, express predominantly cytosolic activities, and are capable of phosphorylating a range of endogenous and non-endogenous sphingoid base substrates. The two genes demonstrate overlapping but distinct temporal and spatial expression patterns in the Drosophila embryo, and timing of expression is consistent with observed changes in long chain base levels throughout development. A null Sk2 transposon insertion mutant demonstrated elevated long chain base levels, impaired flight performance, and diminished ovulation. This is the first reported mutation of a sphingosine kinase in an animal model; the associated phenotypes indicate that Sk1 and Sk2 are not redundant in biological function and that sphingosine kinase is essential for diverse physiological functions in this organism.


Assuntos
Drosophila/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/química , Sequência de Aminoácidos , Animais , Apoptose , Northern Blotting , Divisão Celular , Movimento Celular , Ceramidas/metabolismo , Mapeamento Cromossômico , Clonagem Molecular , DNA Complementar/metabolismo , Proteínas Fúngicas/química , Regulação da Expressão Gênica no Desenvolvimento , Teste de Complementação Genética , Hibridização In Situ , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Ovulação , Fenótipo , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Isoformas de Proteínas , RNA Mensageiro/metabolismo , Reprodução/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Esfingolipídeos/química , Fatores de Tempo
17.
J Lipid Res ; 45(1): 54-62, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-13130120

RESUMO

Sphingolipid metabolites function as signaling molecules in mammalian cells, influencing cell proliferation, migration, and death. Recently, sphingolipid signaling has been implicated in the regulation of developmental processes in Drosophila melanogaster. However, biochemical analysis of endogenous Drosophila sphingoid bases has not been reported. In this study, a rapid HPLC-based method was developed for the analysis of free sphingoid bases endogenous to Drosophila. Four molecular species of endogenous free sphingoid bases were observed in adult flies and identified as C14 and C16 sphingosine (Sph) and C14 and C16 dihydrosphingosine (DHS). The C14 molecular species were the most prevalent, accounting for approximately 94% of the total free sphingoid bases in adult wild-type flies. An Sph kinase (SK) mutant demonstrated significant accumulation of all four sphingoid bases, whereas a serine palmitoyltransferase mutant demonstrated low but detectable levels. When endogenous sphingoid bases were evaluated at different stages of development, the observed ratio of Sph to DHS increased significantly from early embryo to adulthood. Throughout development, this ratio was significantly lower in the SK mutant as compared with the wild-type. This is the first report describing analysis of free C14 and C16 sphingoid bases from Drosophila. The biochemical characterization of these lipids from mutant models of sphingolipid metabolism should greatly facilitate the analysis of the biological significance of these signaling molecules.


Assuntos
Drosophila melanogaster/química , Esfingolipídeos/análise , Esfingolipídeos/química , Animais , Carbono/química , Cromatografia Líquida de Alta Pressão , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Espectrometria de Massas , Esfingolipídeos/metabolismo
18.
J Biol Chem ; 278(25): 22341-9, 2003 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12682045

RESUMO

Sphingolipids are ubiquitous membrane constituents whose metabolites function as signaling molecules in eukaryotic cells. Sphingosine 1-phosphate, a key sphingolipid second messenger, regulates proliferation, motility, invasiveness, and programmed cell death. These effects of sphingosine 1-phosphate and similar phosphorylated sphingoid bases have been observed in organisms as diverse as yeast and humans. Intracellular levels of sphingosine 1-phosphate are tightly regulated by the actions of sphingosine kinase, which is responsible for its synthesis and sphingosine-1-phosphate phosphatase and sphingosine phosphate lyase, the two enzymes responsible for its catabolism. In this study, we describe the cloning of the Caenorhabditis elegans sphingosine phosphate lyase gene along with its functional expression in Saccharomyces cerevisiae. Promoter analysis indicates tissue-specific and developmental regulation of sphingosine phosphate lyase gene expression. Inhibition of C. elegans sphingosine phosphate lyase expression by RNA interference causes accumulation of phosphorylated and unphosphorylated long-chain bases and leads to poor feeding, delayed growth, reproductive abnormalities, and intestinal damage similar to the effects seen with exposure to Bacillus thuringiensis toxin. Our results show that sphingosine phosphate lyase is an essential gene in C. elegans and suggest that the sphingolipid degradative pathway plays a conserved role in regulating animal development.


Assuntos
Caenorhabditis elegans/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Lisofosfolipídeos , Esfingosina/análogos & derivados , Esfingosina/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/crescimento & desenvolvimento , Clonagem Molecular , Primers do DNA , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Esfingosina/química
19.
Development ; 130(11): 2443-53, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12702658

RESUMO

Sphingosine-1-phosphate is a sphingolipid metabolite that regulates cell proliferation, migration and apoptosis through specific signaling pathways. Sphingosine-1-phosphate lyase catalyzes the conversion of sphingosine-1-phosphate to ethanolamine phosphate and a fatty aldehyde. We report the cloning of the Drosophila sphingosine-1-phosphate lyase gene (Sply) and demonstrate its importance for adult muscle development and integrity, reproduction and larval viability. Sply expression is temporally regulated, with onset of expression during mid-embryogenesis. Sply null mutants accumulate both phosphorylated and unphosphorylated sphingoid bases and exhibit semi-lethality, increased apoptosis in developing embryos, diminished egg-laying, and gross pattern abnormalities in dorsal longitudinal flight muscles. These defects are corrected by restoring Sply expression or by introduction of a suppressor mutation that diminishes sphingolipid synthesis and accumulation of sphingolipid intermediates. This is the first demonstration of novel and complex developmental pathologies directly linked to a disruption of sphingolipid catabolism in metazoans.


Assuntos
Aldeído Liases/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Esfingolipídeos/metabolismo , Aldeído Liases/genética , Sequência de Aminoácidos , Animais , Apoptose , Sequência de Bases , Clonagem Molecular , DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Feminino , Fertilidade , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Modelos Biológicos , Dados de Sequência Molecular , Músculo Esquelético/crescimento & desenvolvimento , Mutagênese Insercional , Fenótipo , Homologia de Sequência de Aminoácidos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...