Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Chembiochem ; 19(23): 2438-2442, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30303294

RESUMO

The first biologically relevant clickable probe related to the antitumor marine lipid jaspine B is reported. The concise synthetic route to both enantiomers relied on the supercritical fluid chromatography (SFC) enantiomeric resolution of racemic materials. The eutomeric dextrogyre derivative represents the first jaspine B analogue with enhanced cytotoxicity with IC50 down to 30 nm. These enantiomeric probes revealed a chiralitydependent cytoplasmic imaging of U2OS cancer cells by in situ click labeling.


Assuntos
Alcinos/química , Antineoplásicos/química , Corantes Fluorescentes/química , Sondas Moleculares/química , Esfingosina/análogos & derivados , Alcinos/síntese química , Alcinos/toxicidade , Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Química Click , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/toxicidade , Humanos , Sondas Moleculares/síntese química , Sondas Moleculares/toxicidade , Esfingosina/síntese química , Esfingosina/toxicidade , Estereoisomerismo
2.
Neuropharmacology ; 110(Pt A): 211-222, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27016021

RESUMO

1-Deoxysphingolipids (1-deoxySL) are atypical and neurotoxic sphingolipids formed by alternate substrate usage of the enzyme serine-palmitoyltransferase. Pathologically increased 1-deoxySL formation causes hereditary sensory and autosomal neuropathy type 1 (HSAN1) - a progressive peripheral axonopathy. However, the underlying molecular mechanisms by which 1-deoxySL acts are unknown. Herein we studied the effect of 1-deoxysphinganine (1-deoxySA) and its canonical counterpart sphinganine (SA) in aged cultured neurons comparing their outcome on cell survival and cytoskeleton integrity. 1-deoxySA caused rapid neuronal cytoskeleton disruption and modulated important cytoskeletal regulatory and associated components including Rac1, Ezrin and insulin receptor substrate 53. We show that 1-deoxySA is internalized and metabolized downstream to 1-deoxydihydroceramide since inhibition of ceramide synthase protected neurons from 1-deoxySA-mediated cell death. In addition, 1-deoxySA reduced protein levels of N-methyl-d-aspartate receptor (NMDAR) subunit GluN2B, the postsynaptic density protein 95 and induced cleavage of p35 to p25. Notably, blocking NMDAR activation by MK-801 or memantine significantly prevented 1-deoxySA neurotoxicity. Functional studies of differentiating primary neurons via the patch-clamp technique demonstrated that 1-deoxySA irreversibly depolarizes the neuronal membrane potential in an age-dependent manner. Notably, only neuronal cells that displayed functional NMDAR- and NMDA-induced whole-cell currents responded to 1-deoxySA treatment. Furthermore, pre-exposure to the non-competitive antagonist MK-801 blocked the current response of NMDA and glycine, as well as 1-deoxySA. We conclude that 1-deoxySA-induced neurotoxicity compromises cytoskeletal stability and targets NMDAR signaling in an age-dependent manner. Thus stabilization of cytoskeletal structures and/or inhibition of glutamate receptors could be a potential therapeutic approach to prevent 1-deoxySA-induced neurodegeneration.


Assuntos
Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Esfingolipídeos/toxicidade , Animais , Linhagem Celular Tumoral , Córtex Cerebral , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Proteína 4 Homóloga a Disks-Large/metabolismo , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Antagonistas de Aminoácidos Excitatórios/farmacologia , Glicina/administração & dosagem , Glicina/metabolismo , Humanos , Memantina/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos Endogâmicos C57BL , N-Metilaspartato/administração & dosagem , N-Metilaspartato/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/toxicidade , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/metabolismo
3.
Toxicol Sci ; 150(1): 161-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26719367

RESUMO

FTY720 (fingolimod) is a U.S. Food and Drug Administration-approved drug to treat relapsing remitting multiple sclerosis. FTY720 treatment in pregnant inbred LM/Bc mice results in approximately 60% of embryos having a neural tube defect (NTD). Sphingosine kinases (Sphk1, Sphk2) phosphorylate FTY720 in vivo to form the bioactive metabolite FTY720-1-phosphate (FTY720-P). Cytoplasmic FTY720-P is an agonist for 4 of the 5 sphingosine-1-phosphate (S1P) receptors (S1P1, 3-5) and can also act as a functional antagonist of S1P1, whereas FTY720-P generated in the nucleus inhibits histone deacetylases (HDACs), leading to increased histone acetylation. This study demonstrates that treatment of LM/Bc mouse embryonic fibroblasts (MEFs) with FTY720 results in a significant accumulation of FTY720-P in both the cytoplasmic and nuclear compartments. Elevated nuclear FTY720-P is associated with decreased HDAC activity and increased histone acetylation at H3K18 and H3K23 in LM/Bc MEFs. Treatment of LM/Bc MEFs with FTY720 and a selective Sphk2 inhibitor, ABC294640, significantly reduces the amount of FTY720-P that accumulates in the nucleus. The data provide insight into the relative amounts of FTY720-P generated in the nuclear versus cytoplasmic subcellular compartments after FTY720 treatment and the specific Sphk isoforms involved. The results of this study suggest that FTY720-induced NTDs may involve multiple mechanisms, including: (1) sustained and/or altered S1P receptor activation and signaling by FTY720-P produced in the cytoplasm and (2) HDAC inhibition and histone hyperacetylation by FTY720-P generated in the nucleus that could lead to epigenetic changes in gene regulation.


Assuntos
Núcleo Celular/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Defeitos do Tubo Neural/induzido quimicamente , Organofosfatos/toxicidade , Esfingosina/análogos & derivados , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Citoplasma/metabolismo , Fibroblastos/metabolismo , Histona Desacetilases/metabolismo , Histonas/efeitos dos fármacos , Camundongos Endogâmicos , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/metabolismo , Organofosfatos/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Esfingosina/metabolismo , Esfingosina/toxicidade
4.
Mediators Inflamm ; 2015: 492659, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26576074

RESUMO

Initial and recurrent stroke produces central nervous system (CNS) damage, involving neuroinflammation. Receptor-mediated S1P signaling can influence neuroinflammation and has been implicated in cerebral ischemia through effects on the immune system. However, S1P-mediated events also occur within the brain itself where its roles during stroke have been less well studied. Here we investigated the involvement of S1P signaling in initial and recurrent stroke by using a transient middle cerebral artery occlusion/reperfusion (M/R) model combined with analyses of S1P signaling. Gene expression for S1P receptors and involved enzymes was altered during M/R, supporting changes in S1P signaling. Direct S1P microinjection into the normal CNS induced neuroglial activation, implicating S1P-initiated neuroinflammatory responses that resembled CNS changes seen during initial M/R challenge. Moreover, S1P microinjection combined with M/R potentiated brain damage, approximating a model for recurrent stroke dependent on S1P and suggesting that reduction in S1P signaling could ameliorate stroke damage. Delivery of FTY720 that removes S1P signaling with chronic exposure reduced damage in both initial and S1P-potentiated M/R-challenged brain, while reducing stroke markers like TNF-α. These results implicate direct S1P CNS signaling in the etiology of initial and recurrent stroke that can be therapeutically accessed by S1P modulators acting within the brain.


Assuntos
Isquemia Encefálica/prevenção & controle , Lisofosfolipídeos/fisiologia , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Esfingosina/análogos & derivados , Acidente Vascular Cerebral/prevenção & controle , Animais , Barreira Hematoencefálica , Isquemia Encefálica/etiologia , Cloridrato de Fingolimode/farmacologia , Lisofosfolipídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos ICR , Microinjeções , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Transdução de Sinais , Esfingosina/fisiologia , Esfingosina/toxicidade , Acidente Vascular Cerebral/etiologia , Fator de Necrose Tumoral alfa/análise
5.
Org Biomol Chem ; 13(30): 8241-50, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26133669

RESUMO

Stereoselectivities of electrophilic additions of molecular iodine to enantiomerically pure highly functionalized allylic alcohols with internal nucleophiles have been investigated. The intramolecular nucleophilic attack on the I2-π complex by an oxygen nucleophile to obtain tri- and tetrasubstituted THFs is highly regio-, stereoselective and substrate controlled. The application of this study has been shown by utilizing one of the THFs 4a as a key intermediate to complete the total synthesis of marine anti-cancer natural product 2-epi jaspine B.


Assuntos
Éteres/química , Iodetos/química , Propanóis/química , Esfingosina/análogos & derivados , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclização , Humanos , Concentração Inibidora 50 , Espectroscopia de Ressonância Magnética , Conformação Molecular , Esfingosina/síntese química , Esfingosina/química , Esfingosina/farmacologia , Esfingosina/toxicidade , Estereoisomerismo
6.
Neurosci Lett ; 603: 71-6, 2015 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-26222260

RESUMO

Parkinson's disease (PD) is the second most frequent neurodegenerative disorder. It is characterized by selective degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Early-onset familial forms of PD are associated with mutations in several genes, including parkin, pink1 and dj-1. DJ-1 encodes a protein whose neuroprotective function has not been completely clarified yet. We aim to understand the neuroprotective mechanisms of DJ-1, in particular, DJ-1's involvement in the regulation of the PI3K/PTEN/AKT/mTOR pathway and neuronal autophagy in a neurotoxic context induced by C2-ceramide, by using CAD cells, a murine cathecolaminergic cell line. We demonstrated that C2-ceramide induces CAD cell death associated with decreased phosphorylation of PTEN at Ser380, AKT at Ser473, and mTOR at Ser2448; and increased of autophagic flux (increased LC3-II and autophagosome formation). Additionally, we showed that overexpression of DJ-1 protects against C2-ceramide-induced neuronal death and it is not associated with change in the phosphorylation of mTOR at Ser2448. In conclusion, these data suggest that DJ-1 reinforces the PI3K/AKT survival pathway and inhibits autophagy, probably by a mechanism independent from mTOR.


Assuntos
Autofagia , Neurônios/citologia , Proteínas Oncogênicas/metabolismo , Peroxirredoxinas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Esfingosina/análogos & derivados , Animais , Linhagem Celular , Sobrevivência Celular , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteína Desglicase DJ-1 , Transdução de Sinais , Esfingosina/metabolismo , Esfingosina/toxicidade , Serina-Treonina Quinases TOR/metabolismo
7.
J Biol Chem ; 290(25): 15380-15389, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-25947379

RESUMO

The traditional backbones of mammalian sphingolipids are 2-amino, 1,3-diols made by serine palmitoyltransferase (SPT). Many organisms additionally produce non-traditional, cytotoxic 1-deoxysphingoid bases and, surprisingly, mammalian SPT biosynthesizes some of them, too (e.g. 1-deoxysphinganine from L-alanine). These are rapidly N-acylated to 1-deoxy-"ceramides" with very uncommon biophysical properties. The functions of 1-deoxysphingolipids are not known, but they are certainly dangerous as contributors to sensory and autonomic neuropathies when elevated by inherited SPT mutations, and they are noticeable in diabetes, non-alcoholic steatohepatitis, serine deficiencies, and other diseases. As components of food as well as endogenously produced, these substances are mysteries within an enigma.


Assuntos
Diabetes Mellitus , Mutação , Hepatopatia Gordurosa não Alcoólica , Doenças do Sistema Nervoso Periférico , Serina C-Palmitoiltransferase , Esfingosina/análogos & derivados , Alanina/genética , Alanina/metabolismo , Animais , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Humanos , Lipídeos , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Serina C-Palmitoiltransferase/genética , Serina C-Palmitoiltransferase/metabolismo , Esfingosina/genética , Esfingosina/metabolismo , Esfingosina/toxicidade
8.
Toxicol Lett ; 237(1): 21-9, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26005054

RESUMO

Long-chain bases are present in the oral cavity. Previously we determined that sphingosine, dihydrosphingosine, and phytosphingosine have potent antimicrobial activity against oral pathogens. Here, we determined the cytotoxicities of long-chain bases for oral cells, an important step in considering their potential as antimicrobial agents for oral infections. This information would clearly help in establishing prophylactic or therapeutic doses. To assess this, human oral gingival epithelial (GE) keratinocytes, oral gingival fibroblasts (GF), and dendritic cells (DC) were exposed to 10.0-640.0 µM long-chain bases and glycerol monolaurate (GML). The effects of long-chain bases on cell metabolism (conversion of resazurin to resorufin), membrane permeability (uptake of propidium iodide or SYTOX-Green), release of cellular contents (LDH), and cell morphology (confocal microscopy) were all determined. GE keratinocytes were more resistant to long-chain bases as compared to GF and DC, which were more susceptible. For DC, 0.2-10.0 µM long-chain bases and GML were not cytotoxic; 40.0-80.0 µM long-chain bases, but not GML, were cytotoxic; and 80.0 µM long-chain bases induced cellular damage and death in less than 20 min. The LD50 of long-chain bases for GE keratinocytes, GF, and DC were considerably higher than their minimal inhibitory concentrations for oral pathogens, a finding important to pursuing their future potential in treating periodontal and oral infections.


Assuntos
Células Dendríticas/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Gengiva/citologia , Queratinócitos/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/toxicidade , Anti-Infecciosos/toxicidade , Diferenciação Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Células Dendríticas/metabolismo , Fibroblastos/metabolismo , Gengiva/efeitos dos fármacos , Humanos , Queratinócitos/metabolismo , Dose Letal Mediana , Saliva/química
9.
Brain Res ; 1620: 17-28, 2015 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25998538

RESUMO

α-Crystallin with two isoforms, αA-crystallin (HSPB4) and αB-crystallin (HSPB5), is found in eye lens, spleen, lung, kidney, cornea, skin, but also in brain. Several studies revealed roles of αA/αB-crystallin in regulating cell viability and protection in the central nervous system. We previously demonstrated that α-crystallin serves as an intracellular protectant in astrocytes. Compared to well-studied intracellular functions of α-crystallin, there is limited proof for the role of α-crystallin as extracellular protectant. In order to clarify protective effects of extracellular αA/αB-crystallin, we exposed astrocytes to the toxic agents, staurosporine or C2-ceramide, or serum-starvation in the presence of αA/αB-crystallin. Extracellular αA/αB-crystallin protected astrocytes from staurosporine- and C2-ceramide-induced cell death. In addition, extracellular αB-crystallin/HSPB5 effectively promoted astrocytes viability through phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and extracellular signal-regulated kinase 1/2 (ERK1/2), p38 mitogen-activated protein kinases (p38) and c-Jun N-terminal kinases (JNK) signaling pathways under serum-deprivation. Furthermore, αB-crystallin/HSPB5 decreases the staurosporine-mediated cleavage of caspase 3 through PI3K/Akt signaling preventing apoptosis of astrocytes. Thus, the current study indicates that extracellular αA/αB-crystallin protects astrocytes exposed to various harmful stimuli. Furthermore, application of αB-crystallin/HSPB5 to isolated rat brain mitochondria inhibits ROS generation induced by complex III inhibition with Antimycin A.


Assuntos
Astrócitos/fisiologia , Morte Celular/fisiologia , Espaço Extracelular/metabolismo , Mitocôndrias/metabolismo , alfa-Cristalinas/metabolismo , Animais , Antimicina A/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Escherichia coli , Espaço Extracelular/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Mitocôndrias/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Esfingosina/análogos & derivados , Esfingosina/toxicidade , Estaurosporina/toxicidade , Serina-Treonina Quinases TOR/metabolismo , alfa-Cristalinas/administração & dosagem
10.
Int J Dev Neurosci ; 40: 24-34, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25450526

RESUMO

Several chemotherapeutic drugs are known to cause neurotoxicity. Platinum-based agents in use or in clinical trials display neurotoxic potential accompanied by neurological complications; recent studies have identified a large number of behavioural issues in paediatric oncology patients. To understand the toxicity of platinum drugs at the molecular and cellular levels, this study compares the possible cytotoxic effects of an older platinum compound, cisplatin and a new platinum compound, [Pt(O,O'-acac)(γ-acac)(DMS)], on the CNS of postnatally developing rats, which is much more vulnerable to injury than the CNS of adult rats. Since several drugs interact with neurotransmitters during neuronal maturation, we performed immunostainings with antibodies raised against markers of glutamate and GABA, the major neurotransmitters in the cerebellum. After a single injection of cisplatin at postnatal day 10 (PD10), the labelling of Purkinje cells with the neurotransmitter markers evidenced alterations between PD11 and PD30, i.e. atrophy of the dendrite tree, changes in the distribution of synaptic contacts of parallel and climbing fibres, delay in the elimination of transient synapses on cell soma and severely impaired pinceau formation at the axon hillock. After treatment with [Pt(O,O'-acac)(γ-acac)(DMS)], the sole relevant change concerned the timing of climbing fibres elimination; the transient synapses disappearance on the Purkinje cell soma was delayed in some cells; instead, the growth of Purkinje cell dendrite tree was normal as was the formation of inhibitory synaptic contacts on these neurons. These findings add new evidence not only on the lower neurotoxicity of [Pt(O,O'-acac)(γ-acac)(DMS)] vs cisplatin but also on the involvement of neurotransmitters and relative synaptic connections in the maturation of central nerve tissue.


Assuntos
Cerebelo , Cisplatino/toxicidade , Síndromes Neurotóxicas/etiologia , Neurotoxinas/toxicidade , Esfingosina/análogos & derivados , Transmissão Sináptica/efeitos dos fármacos , Fatores Etários , Animais , Animais Recém-Nascidos , Calbindinas/metabolismo , Cerebelo/efeitos dos fármacos , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glutamato Descarboxilase/metabolismo , Ratos , Ratos Wistar , Receptores de Glutamato/metabolismo , Esfingosina/toxicidade , Proteína Vesicular 1 de Transporte de Glutamato
11.
J Mol Neurosci ; 54(4): 698-703, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25239520

RESUMO

FTY720 (fingolimod), the sphingosine-1-phosphate (S1P) analogue, has been experimentally indicated to exert substantial ameliorating effects in animal models of Alzheimer's disease (AD). The present work aims to answer whether central S1P receptor 1 (S1P1) plays significant role in the impact of fingolimod in AD. To verify the prominence of central FTY720 phosphorylation, DMS (sphingosine kinase inhibitor) was infused intracerebrally in parallel with systemic FTY720 administration to prevent central formation of FTY720-P as the recognized active ligand for S1PRs. The corresponding S1P1 modulation was also investigated using the pharmacological blockage of central S1P1 by W123. Both DMS and W123 were efficiently capable of suppressing FTY720-ameliorating effects in AD animals, either on memory deficit or on COX-II and TNF-α expression. Our data conclude that experimental benefits of FTY720 in the context of AD depend on central S1P1 modulation, as well as on S1P kinase activity in the brain.


Assuntos
Doença de Alzheimer/metabolismo , Propilenoglicóis/toxicidade , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Doença de Alzheimer/etiologia , Doença de Alzheimer/fisiopatologia , Animais , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Inibidores Enzimáticos/farmacologia , Cloridrato de Fingolimode , Masculino , Aprendizagem em Labirinto , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Ratos , Ratos Wistar , Esfingosina/farmacologia , Esfingosina/toxicidade , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
Neurotoxicology ; 44: 140-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24792327

RESUMO

It is now well established that mitochondria are organelles that, far from being static, are subject to a constant process of change. This process, which has been called mitochondrial dynamics, includes processes of both fusion and fission. Loss of Pink1 (PTEN-induced putative kinase 1) function is associated with early onset recessive Parkinson's disease and it has been proposed that mitochondrial dynamics might be affected by loss of the mitochondrial kinase. Here, we report the effects of silencing Pink1 on mitochondrial fusion and fission events in dopaminergic neuron cell lines. Cells lacking Pink1 were more sensitive to cell death induced by C2-Ceramide, which inhibits proliferation and induces apoptosis. In the same cell lines, mitochondrial morphology was fragmented and this was enhanced by application of forskolin, which stimulates the cAMP pathway that phosphorylates Drp1 and thereby inactivates it. Cells lacking Pink1 had lower Drp1 and Mfn2 expression. Based on these data, we propose that Pink1 may exert a neuroprotective role in part by limiting mitochondrial fission.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas Quinases/fisiologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Neurônios Dopaminérgicos/efeitos dos fármacos , Regulação para Baixo , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Rotenona/toxicidade , Esfingosina/análogos & derivados , Esfingosina/toxicidade
13.
Int J Mol Sci ; 15(2): 2660-71, 2014 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-24549171

RESUMO

Safingol, a L-threo-dihydrosphingosine, induced the nuclear translocation of a mitochondrial apoptogenic mediator--endonuclease G (endo G)--and apoptosis of human oral squamous cell carcinoma (SCC) cells. Upstream mediators remain largely unknown. The levels of hydrogen peroxide (H2O2) in cultured oral SCC cells were measured. Treatment with safingol increased intracellular H2O2 levels but not extracellular H2O2 levels, indicating the production of H2O2. The cell killing effect of safingol and H2O2 was diminished in the presence of reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC). Dual staining of cells with annexin V and propidium iodide (PI) revealed that apoptotic cell death occurred by treatment with H2O2 and safingol. The number of apoptotic cells was reduced in the presence of NAC. In untreated cells, endo G distributed in the cytoplasm and an association of endo G with mitochondria was observed. After treatment with H2O2 and safingol, endo G was distributed to the nucleus and cytoplasm, indicating the nuclear translocation of the mitochondrial factor. NAC prevented the increase of apoptotic cells and the translocation of endo G. Knock down of endo G diminished the cell killing effect of H2O2 and safingol. These results suggest that H2O2 is involved in the endo G-mediated apoptosis of oral SCC cells by safingol.


Assuntos
Apoptose/efeitos dos fármacos , Endodesoxirribonucleases/metabolismo , Peróxido de Hidrogênio/toxicidade , Esfingosina/análogos & derivados , Acetilcisteína/farmacologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Endodesoxirribonucleases/antagonistas & inibidores , Endodesoxirribonucleases/genética , Humanos , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Esfingosina/toxicidade
14.
Int J Mol Sci ; 15(2): 3336-55, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24566153

RESUMO

Ceramides are second messengers involved in several intracellular processes in cancer cells, amongst others. The aim of this study was to evaluate the anti-tumor efficacy of C2-ceramide (C2-Cer; N-acetyl-D-sphingosine) by investigating cell death and autophagy in head and neck squamous cell carcinoma (HNSCC) cells. C2-Cer showed concentration-dependent cytotoxicity in HN4 and HN30 cell lines. It simultaneously induced caspase-3-independent apoptosis and programmed necrosis. C2-Cer markedly increased the expression level of microtubule-associated protein 1 light chain 3B (LC3B) type II associated with protective autophagy. An autophagy inhibitor enhanced C2-Cer-mediated cytotoxicity, while a programmed-necrosis inhibitor produced the opposite effect. Furthermore, C2-Cer up-regulated the phosphorylation of extracellular signal-regulated kinase 1/2, but down-regulated its downstream substrate phospho-mammalian target of rapamycin (p-mTOR) during the autophagy process. These results suggested that C2-Cer exerts anti-tumor effects by inducing programmed apoptosis and necrosis in HNSCC, and these cytotoxic effects are enhanced by an autophagy inhibitor.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Esfingosina/análogos & derivados , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Fragmentação do DNA/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Necrose , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Esfingosina/toxicidade , Serina-Treonina Quinases TOR/metabolismo
15.
Mol Plant Microbe Interact ; 26(8): 868-79, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23617414

RESUMO

To investigate plant programmed cell death (PCD), we developed the model system using phytotoxin AAL, which is produced by necrotrophic pathogen Alternaria alternata f. sp. lycopersici, and AAL-sensitive Nicotiana umbratica. We previously reported that ethylene (ET) signaling plays a pivotal role in AAL-triggered cell death (ACD). However, downstream signaling of ET to ACD remains unclear. Here, we show that the modulator of AAL cell death 1 (MACD1), which is an APETALA2/ET response factor (ERF) transcription factor, participates in ACD and acts downstream of ET signaling during ACD. MACD1 is a transcriptional activator and MACD1 overexpression plants showed earlier ACD induction than control plants, suggesting that MACD1 positively regulates factors affecting cell death. To investigate the role of MACD1 in PCD, we used Arabidopsis thaliana and a structural analog of AAL, fumonisin B1 (FB1). FB1-triggered cell death was compromised in ET signaling and erf102 mutants. The loh2 mutants showed sensitivity to AAL, and the loh2-1/erf102 double mutant compromised ACD, indicating that ERF102 also participates in ACD. To investigate the PCD-associated genes regulated by ERF102, we compared our microarray data using ERF102 overexpression plants with the database of upregulated genes by AAL treatment in loh2 mutants, and found genes under the control of ERF102 in ACD.


Assuntos
Apoptose/efeitos dos fármacos , Nicotiana/citologia , Nicotiana/efeitos dos fármacos , Proteínas de Plantas/metabolismo , Esfingosina/toxicidade , Apoptose/fisiologia , Etilenos/metabolismo , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Regulação da Expressão Gênica de Plantas/fisiologia , Inativação Gênica , Proteínas de Plantas/genética , Transdução de Sinais
16.
Atherosclerosis ; 225(2): 315-21, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23084711

RESUMO

OBJECTIVE: The kinase receptor-interacting protein (RIP) 1, a serine/threonine protein kinase, is a key signaling molecule for necrosis. The possible involvement of RIP1 in palmitate-induced macrophage death and its underlying molecular mechanism was investigated in this study. METHODS: Cell viability was measured by an MTT reduction assay. The type of cell death was determined by staining with annexin V, propidium iodide (PI) and the APOPercentage dye, and by examining cell morphology using transmission electron microscopy. The down-regulation of RIP1 was performed by siRNA transfection. Intracellular reactive oxygen species (ROS) were measured by staining with H(2)DCF-DA. RESULTS: Palmitate largely induced necrosis in RAW 264.7 cells, whereas C2-ceramide induced apoptosis. Palmitate-induced necrosis was inhibited by Necrostatin-1, an inhibitor of RIP1, and by RIP1 siRNA transfection, whereas ordinary cell death was not inhibited by z-VAD-fmk. In addition, the presence of palmitate caused a significant increase in intracellular ROS levels compared to control cells. Pre-treatment with Tempol, a cell permeable ROS scavenger, and MnTBAP, an inhibitor of mitochondrial oxidative stress, protected cells from palmitate-induced cell death. Furthermore, the down-regulation of RIP1 by siRNA transfection significantly decreased palmitate-induced ROS generation compared to control cells. CONCLUSION: The findings reported herein indicate that palmitate induces necrotic cell death via RIP1-dependent ROS generation in RAW 264.7 cells. These findings may provide a new mechanism that explains the link between elevated levels of free fatty acids (FFAs) and macrophage death.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Macrófagos/efeitos dos fármacos , Ácido Palmítico/toxicidade , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Óxidos N-Cíclicos/farmacologia , Relação Dose-Resposta a Droga , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Proteínas Ativadoras de GTPase/genética , Imidazóis/farmacologia , Indóis/farmacologia , Macrófagos/enzimologia , Macrófagos/ultraestrutura , Metaloporfirinas/farmacologia , Camundongos , Microscopia Eletrônica de Transmissão , Necrose , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Esfingosina/análogos & derivados , Esfingosina/toxicidade , Marcadores de Spin , Transfecção
17.
Int J Pharm ; 434(1-2): 191-8, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22643227

RESUMO

Phytosphingosine and methyl derivatives are important mediators on cellular processes, and are associated with cell growth and death. The antitumor activity of N,N,N-trimethylphytosphingosine-iodide (TMP) as a novel potent inhibitor of angiogenesis and metastasis was evaluated in B16F10 murine melanoma cells. The results indicated that TMP itself effectively inhibited in vitro cell migration, tube formation, and the expression of angiogenic factors as well as in vivo lung metastasis. However, TMP slightly suppressed in vivo experimental tumor metastasis in its free form and induced side effects including hemolysis and local side effects. Therefore, in an attempt to reduce the toxicity and the undesirable side effects of TMP, a liposomal formulation was prepared and tested for its effectiveness. TMP liposomes retained the effectiveness of TMP in vitro while side effects were reduced, and both in vivo experimental and spontaneous tumor metastasis were significantly suppressed. These results support the conclusion that TMP effectively inhibits in vitro angiogenesis as well as in vivo metastasis, and a liposomal formulation is more efficient delivery system for TMP treatment than solution.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Melanoma Experimental/tratamento farmacológico , Compostos de Amônio Quaternário/farmacologia , Esfingosina/análogos & derivados , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/toxicidade , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Humanos , Lipossomos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Compostos de Amônio Quaternário/administração & dosagem , Compostos de Amônio Quaternário/toxicidade , Ratos , Esfingosina/administração & dosagem , Esfingosina/farmacologia , Esfingosina/toxicidade
18.
J Mol Neurosci ; 47(3): 582-94, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22212487

RESUMO

The etiology of Parkinson's disease (PD) remains unknown. Mutations in several genes, including PINK1, have provided an understanding of the molecular mechanisms of this pathology. We analyzed the role of PINK1 overexpression (wild-type PINK1 or PINK1 with G309D or L347P mutations) on neurotoxicity associated with C2-ceramide exposure in CAD cells. CAD cells were transiently transfected with either PINK1 (wild type or mutated) or with empty vector and then treated with 25-µM C2-ceramide for 6 h. Cell viability and mitochondrial membrane potential were analyzed by flow cytometry, expression of Bax and Bcl-2 was determined by real-time PCR, and AKT phosphorylation was analyzed by western blot. CAD cells overexpressing wild-type PINK1 and treated with C2-ceramide showed lower percentages of depolarized mitochondria, lower expressions of Bax and higher expressions of Bcl-2 than non-transfected cells. In addition, wild-type PINK1 rescued C2-ceramide-induced inhibition of AKT phosphorylation. Overexpression of PINK1 G309D mutation caused an increase of depolarized mitochondria, a decrease of Bax and an increase in Bcl-2 expression levels. PINK1 L4347P mutation was associated with a higher drop in mitochondrial membrane potential and increased expression of Bax, with minimal variation in the expression of Bcl-2. PINK1 mutations did not result in variations of AKT phosphorylation. We suggest that by preventing mitochondrial dysfunction and reinforcing anti-apoptotic and neuronal survival pathways such as Bcl-2 and PI3K/AKT, PINK1 confers a neuroprotective effect against the neurotoxin C2-ceramide. These effects were abrogated by PINK1 mutations.


Assuntos
Doença de Parkinson/genética , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Quinases/genética , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Esfingosina/análogos & derivados , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular , Sobrevivência Celular/genética , Humanos , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/patologia , Doença de Parkinson/enzimologia , Doença de Parkinson/patologia , Proteínas Quinases/biossíntese , Transdução de Sinais/efeitos dos fármacos , Esfingosina/antagonistas & inibidores , Esfingosina/toxicidade
19.
BMC Musculoskelet Disord ; 12: 279, 2011 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-22151889

RESUMO

BACKGROUND: FTY720 (Fingolimod) is a novel immunosuppressive drug investigated in clinical trials for organ transplantation and multiple sclerosis. It acts as a functional sphingosine-1-phosphate (S1P) receptor antagonist, thereby inhibiting the egress of lymphocytes from secondary lymphoid organs. As S1P is able to prevent IL-1beta induced cartilage degradation, we examined the direct impact of FTY720 on cytokine induced cartilage destruction. METHODS: Bovine chondrocytes were treated with the bioactive phosphorylated form of FTY720 (FTY720-P) in combination with IL-1beta or TNF-alpha. Expression of MMP-1,-3.-13, iNOS and ADAMTS-4,-5 and COX-2 was evaluated using quantitative real-time PCR and western blot. Glycosaminoglycan depletion from cartilage explants was determined using a 1,9-dimethylene blue assay and safranin O staining. RESULTS: FTY720-P significantly reduced IL-1beta and TNF-alpha induced expression of iNOS. In contrast FTY720-P increased MMP-3 and ADAMTS-5 mRNA expression. Furthermore depletion of glycosaminoglycan from cartilage explants by IL-1beta and TNF-alpha was significantly enhanced by FTY720-P in an MMP-3 dependent manner. CONCLUSIONS: Our results suggest that FTY720 may enhance cartilage degradation in pro-inflammatory environment.


Assuntos
Cartilagem Articular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Glicosaminoglicanos/antagonistas & inibidores , Glicosaminoglicanos/metabolismo , Imunossupressores/toxicidade , Propilenoglicóis/toxicidade , Esfingosina/análogos & derivados , Animais , Cartilagem Articular/enzimologia , Cartilagem Articular/metabolismo , Bovinos , Células Cultivadas , Condrócitos/metabolismo , Condrócitos/patologia , Cloridrato de Fingolimode , Interleucina-1beta/fisiologia , Metaloproteinase 13 da Matriz/biossíntese , Metaloproteinase 3 da Matriz/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Esfingosina/toxicidade , Fator de Necrose Tumoral alfa/fisiologia
20.
J Biomed Sci ; 18: 55, 2011 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-21838918

RESUMO

BACKGROUND: Sphingosylphosphorylcholine (SPC) acts as a potent lipid mediator and signaling molecule in various cell types. In the present study, we investigated the effects of SPC on melanogenesis and SPC-modulated signaling pathways related to melanin synthesis. METHODS: Melanin production was measured in Mel-Ab cells. A luciferase assay was used to detect transcriptional activity of the MITF promoter. Western blot analysis was performed to examine SPC-induced signaling pathways. RESULTS: SPC produced significant hypopigmentation effects in a dose-dependent manner. It was found that SPC induced not only activation of Akt but also stimulation of mTOR, a downstream mediator of the Akt signaling pathway. Moreover, SPC decreased the levels of LC3 II, which is known to be regulated by mTOR. Treatment with the mTOR inhibitor rapamycin eliminated decreases in melanin and LC3 II levels by SPC. Furthermore, we found that the Akt inhibitor LY294002 restored SPC-mediated downregulation of LC3 II and inhibited the activation of mTOR by SPC. CONCLUSIONS: Our data suggest that the mTOR signaling pathway is involved in SPC-modulated melanin synthesis.


Assuntos
Hipopigmentação/metabolismo , Melaninas/biossíntese , Fosforilcolina/análogos & derivados , Transdução de Sinais/fisiologia , Esfingosina/análogos & derivados , Serina-Treonina Quinases TOR/metabolismo , Análise de Variância , Animais , Western Blotting , Linhagem Celular , Cromonas/farmacologia , Relação Dose-Resposta a Droga , Hipopigmentação/induzido quimicamente , Luciferases , Camundongos , Microscopia de Contraste de Fase , Monofenol Mono-Oxigenase/metabolismo , Morfolinas/farmacologia , Fosforilcolina/farmacologia , Fosforilcolina/toxicidade , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sirolimo/farmacologia , Esfingosina/farmacologia , Esfingosina/toxicidade , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA