Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Biol Chem ; 290(5): 3057-68, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25492865

RESUMO

Apurinic/apyrimidinic endonuclease/redox factor-1 (APE1/Ref-1) (henceforth referred to as Ref-1) is a multifunctional protein that in addition to its base excision DNA repair activity exerts redox control of multiple transcription factors, including nuclear factor κ-light chain enhancer of activated B cells (NF-κB), STAT3, activator protein-1 (AP-1), hypoxia-inducible factor-1 (HIF-1), and tumor protein 53 (p53). In recent years, Ref-1 has emerged as a promising therapeutic target in cancer, particularly in pancreatic ductal carcinoma. Although a significant amount of research has centered on Ref-1, no wide-ranging approach had been performed on the effects of Ref-1 inhibition and transcription factor activity perturbation. Starting with a broader approach, we identified a previously unsuspected effect on the nuclear factor erythroid-related factor 2 (NRF2), a critical regulator of cellular defenses against oxidative stress. Based on genetic and small molecule inhibitor-based methodologies, we demonstrated that repression of Ref-1 potently activates NRF2 and its downstream targets in a dose-dependent fashion, and that the redox, rather than the DNA repair function of Ref-1 is critical for this effect. Intriguingly, our results also indicate that this pathway does not involve reactive oxygen species. The link between Ref-1 and NRF2 appears to be present in all cells tested in vitro, noncancerous and cancerous, including patient-derived tumor samples. In particular, we focused on understanding the implications of the novel interaction between these two pathways in primary pancreatic ductal adenocarcinoma tumor cells and provide the first evidence that this mechanism has implications for overcoming the resistance against experimental drugs targeting Ref-1 activity, with clear translational implications.


Assuntos
DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Neoplasias Pancreáticas/metabolismo , Linhagem Celular Tumoral , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Humanos , Fator 2 Relacionado a NF-E2/genética , Oxirredução , Neoplasias Pancreáticas/genética , Espécies Reativas de Oxigênio/metabolismo
2.
Traffic ; 11(5): 601-15, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20412078

RESUMO

Different primary lysosomal trafficking defects lead to common alterations in lipid trafficking, suggesting cooperative interactions among lysosomal lipids. However, cellular analysis of the functional consequences of this phenomenon is lacking. As a test case, we studied cells with defective Niemann-Pick C1 (NPC1) protein, a cholesterol trafficking protein whose defect gives rise to lysosomal accumulation of cholesterol and other lipids, leading to NPC disease. NPC1 cells also develop a secondary defect in acid sphingomyelinase (SMase) activity despite a normal acid SMase gene (SMPD1). When acid SMase activity was restored to normal levels in NPC1-deficient CHO cells through SMPD1 transfection, there was a dramatic reduction in lysosomal cholesterol. Two other defects, excess lysosomal bis-(monoacylglycerol) phosphate (BMP) and defective transferrin receptor (TfR) recycling, were also markedly improved. To test its relevance in human cells, the acid SMase activity defect in fibroblasts from NPC1 patients was corrected by SMPD1 transfection or acid SMase enzyme replacement. Both treatments resulted in a dramatic reduction in lysosomal cholesterol. These data show that correcting one aspect of a complex lysosomal lipid storage disease can reduce the cellular consequences even if the primary genetic defect is not corrected.


Assuntos
Doenças de Niemann-Pick/genética , Doenças de Niemann-Pick/metabolismo , Proteínas/genética , Proteínas/metabolismo , Animais , Antígenos CD , Células CHO , Colesterol/genética , Colesterol/metabolismo , Cricetinae , Cricetulus , Fibroblastos/metabolismo , Humanos , Lipídeos/genética , Lisossomos/genética , Lisossomos/metabolismo , Doença de Niemann-Pick Tipo C/genética , Doença de Niemann-Pick Tipo C/metabolismo , Transporte Proteico/genética , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/metabolismo , Transfecção
3.
J Biol Chem ; 286(44): 38069-38078, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-21914808

RESUMO

To understand the mechanisms of ceramide-based responses to hypoxia, we performed a mass spectrometry-based survey of ceramide species elicited by a wide range of hypoxic conditions (0.2-5% oxygen). We describe a rapid, time-dependent, marked up-regulation of dihydroceramides (DHCs) in mammalian cells and in the lungs of hypoxic rats. The increase affected all DHC species and was proportional with the depth and duration of hypoxia, ranging from 2- (1 h) to 10-fold (24 h), with complete return to normal after 1 h of reoxygenation at the expense of increased ceramides. We demonstrate that a DHC-based response to hypoxia occurs in a hypoxia-inducible factor-independent fashion and is catalyzed by the DHC desaturase (DEGS) in the de novo ceramide pathway. Both the impact of hypoxia on DHC molecular species and its inhibitory effect on cell proliferation were reproduced by knockdown of DEGS1 or DEGS2 by siRNA during normoxia. Conversely, overexpression of DEGS1 or DEGS2 attenuated the DHC accumulation and increased cell proliferation during hypoxia. Based on the amplitude and kinetics of DHC accumulation, the enzymatic desaturation of DHCs fulfills the criteria of an oxygen sensor across physiological hypoxic conditions, regulating the balance between biologically active components of ceramide metabolism.


Assuntos
Ceramidas/farmacologia , Hipóxia , Oxirredutases/química , Animais , Técnicas Biossensoriais , Linhagem Celular Tumoral , Proliferação de Células , Ceramidas/química , Relação Dose-Resposta a Droga , Humanos , Cinética , Masculino , Espectrometria de Massas/métodos , Camundongos , Oxigênio/química , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima
4.
IUBMB Life ; 63(2): 94-100, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21360638

RESUMO

Multiple studies have consistently established that miR (microRNA)-210 induction is a feature of the hypoxic response in both normal and transformed cells. Here, we discuss the emerging biochemical functions of this miRNA and anticipate potential clinical applications. miR-210 is a robust target of hypoxia-inducible factor, and its overexpression has been detected in a variety of cardiovascular diseases and solid tumors. High levels of miR-210 have been linked to an in vivo hypoxic signature and associated with adverse prognosis in cancer patients. A wide spectrum of miR-210 targets have been identified, with roles in mitochondrial metabolism, angiogenesis, DNA repair, and cell survival. Such targets may broadly affect the evolution of tumors and other pathological settings, such as ischemic disorders. Harnessing the knowledge of miR-210's actions may lead to novel diagnostic and therapeutic approaches.


Assuntos
Biomarcadores/análise , Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , MicroRNAs , Animais , Apoptose , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , Hipóxia Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Reparo do DNA , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Hipóxia/genética , Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Endogâmicos , MicroRNAs/biossíntese , MicroRNAs/genética , Mitocôndrias/metabolismo , Terapia de Alvo Molecular , Neoplasias/genética , Neoplasias/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Estresse Oxidativo , Prognóstico
5.
Nat Cell Biol ; 5(9): 781-92, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12907943

RESUMO

Excess cellular cholesterol induces apoptosis in macrophages, an event likely to promote progression of atherosclerosis. The cellular mechanism of cholesterol-induced apoptosis is unknown but had previously been thought to involve the plasma membrane. Here we report that the unfolded protein response (UPR) in the endoplasmic reticulum is activated in cholesterol-loaded macrophages, resulting in expression of the cell death effector CHOP. Cholesterol loading depletes endoplasmic reticulum calcium stores, an event known to induce the UPR. Furthermore, endoplasmic reticulum calcium depletion, the UPR, caspase-3 activation and apoptosis are markedly inhibited by selective inhibition of cholesterol trafficking to the endoplasmic reticulum, and Chop-/- macrophages are protected from cholesterol-induced apoptosis. We propose that cholesterol trafficking to endoplasmic reticulum membranes, resulting in activation of the CHOP arm of the UPR, is the key signalling step in cholesterol-induced apoptosis in macrophages.


Assuntos
Apoptose/fisiologia , Colesterol/toxicidade , Doença da Artéria Coronariana/metabolismo , Retículo Endoplasmático/metabolismo , Macrófagos/metabolismo , Dobramento de Proteína , Animais , Apoptose/efeitos dos fármacos , Transporte Biológico Ativo/efeitos dos fármacos , Transporte Biológico Ativo/fisiologia , Proteínas Estimuladoras de Ligação a CCAAT/deficiência , Proteínas Estimuladoras de Ligação a CCAAT/genética , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Células Cultivadas , Colesterol/metabolismo , Doença da Artéria Coronariana/fisiopatologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/ultraestrutura , Feminino , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestrutura , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Transcrição CHOP , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
6.
Arterioscler Thromb Vasc Biol ; 28(10): 1723-30, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18669882

RESUMO

OBJECTIVE: The key initial step in atherogenesis is the subendothelial retention of apolipoprotein B-containing lipoproteins. Acid sphingomyelinase (acid SMase), an enzyme present extracellularly within the arterial wall, strongly enhances lipoprotein retention in model systems in vitro, and retained lipoproteins in human plaques are enriched in ceramide, a product of SMase. We now sought to test a direct causative role for acid SMase in lipoprotein retention and atherogenesis in vivo. METHODS AND RESULTS: We studied atherogenesis and lipoprotein retention in Asm(-/-) versus Asm(+/+) mice on the Apoe(-/-) and Ldlr(-/-) backgrounds. Asm(-/-);Apoe(-/-) mice had a approximately 40% to 50% decrease in early foam cell aortic root lesion area compared with Asm(+/+);Apoe(-/-) mice (P<0.05) despite no difference in plasma cholesterol or lipoproteins. To assay lipoprotein retention in vivo, the two groups of mice were injected with fluorescently labeled Apoe(-/-) lipoproteins. Early foam cell lesions of Asm(-/-);Apoe(-/-) mice showed a striking 87% reduction in lipoprotein trapping (P<0.0001) compared with Asm(+/+);Apoe(-/-) lesions. Similar results were obtained with Ldlr(-/-) mice, including an 81% reduction in lipoprotein retention within Asm(-/-);Ldlr(-/-) lesions compared with Asm(+/+);Ldlr(-/-) lesions (P<0.0005). CONCLUSIONS: These findings support a causal role for acid SMase in lipoprotein retention and lesion progression and provides further support for the response-to-retention model of atherogenesis.


Assuntos
Aterosclerose/enzimologia , Lipoproteínas/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Animais , Aorta Torácica/enzimologia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/etiologia , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Gorduras na Dieta , Modelos Animais de Doenças , Progressão da Doença , Feminino , Células Espumosas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Receptores de LDL/genética , Receptores de LDL/metabolismo , Esfingomielina Fosfodiesterase/deficiência , Esfingomielina Fosfodiesterase/genética
7.
Arterioscler Thromb Vasc Biol ; 27(1): 190-6, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17038631

RESUMO

OBJECTIVE: PLTP and apoE play important roles in lipoprotein metabolism and atherosclerosis. It is known that formation of macrophage-derived foam cells (which highly express PLTP and apoE) is the critical step in the process of atherosclerosis. We investigated the relationship between PLTP and apoE in macrophages and the atherogenic relevance in a mouse model. METHODS AND RESULTS: We transplanted PLTP-deficient mouse bone marrow into apoE-deficient mice (PLTP-/- --> apoE-/-), creating a mouse model with PLTP deficiency and apoE expression exclusively in the macrophages. We found that PLTP-/- --> apoE-/- mice have significantly lower PLTP activity, compared with controls (WT --> apoE-/-; 20%, P<0.01). On a Western diet, PLTP-/- --> apoE-/- mice have significantly lower plasma apoE than that of WT --> apoE-/- mice (63%, P<0.001), and PLTP-deficient macrophages secrete significantly less apoE than WT macrophages (44%, P<0.01). Moreover, PLTP-/- --> apoE-/- mice have significantly higher plasma cholesterol (98%, P<0.001) and phospholipid (107%, P<0.001) than that of WT --> apoE-/- mice, thus increasing atherosclerotic lesions in the aortic arch and root (403%, P<0.001), as well as the entire aorta (298%, P<0.001). CONCLUSIONS: Macrophage PLTP deficiency causes a significant reduction of apoE secretion from the cells, and this in turn promotes the accumulation of cholesterol in the circulation and accelerates the development of atherosclerosis.


Assuntos
Apolipoproteínas E/metabolismo , Aterosclerose/sangue , Aterosclerose/etiologia , Colesterol/sangue , Macrófagos/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Animais , Apolipoproteínas E/genética , Aterosclerose/patologia , Transplante de Medula Óssea/patologia , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Suscetibilidade a Doenças , Feminino , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Transferência de Fosfolipídeos/genética , Fosfolipídeos/sangue , Fatores de Risco
8.
Arterioscler Thromb Vasc Biol ; 25(8): 1704-10, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15905467

RESUMO

OBJECTIVE: Humans with high expression of apolipoprotein(a) [apo(a)] and high plasma levels of lipoprotein(a) [Lp(a)] are at increased risk for atherosclerosis, but the mechanism is not known. We have previously shown that the KIV(5-8) domain of apo(a) has unique cell-surface binding properties, and naturally occurring fragments of apo(a) encompassing this domain are thought to be atherogenic in humans. To investigate the effect of KIV(5-8) on lipoprotein metabolism and atherosclerosis in vivo, we created several independent lines of liver-targeted KIV(5-8) transgenic mice. METHODS AND RESULTS: The transgenic mice have plasma apo(a) peptide concentrations that are similar to Lp(a) concentrations in humans at risk for coronary artery disease. Remarkably, the transgenic mice had a 2- to 4-fold increase in cholesterol-rich remnant lipoproteins (RLPs) when fed a cholesterol-rich diet, and a 5- to 20-fold increase in atherosclerosis lesion area in the aortic root. Using an in vivo clearance study, we found only slight differences in the triglyceride and apolipoprotein B secretion rates between the 2 groups of mice, suggesting an RLP clearance defect. Using an isolated perfused mouse liver system, we showed that transgenic livers had a slower rate of RLP removal, which was retarded further when KIV(5-8), full-length apo(a), or Lp(a) were added to the perfusate. An apo(a) peptide that does not interact with cells, K(IV2)3, did not retard RLP removal, and low-density lipoprotein (LDL) had a much smaller effect than Lp(a). CONCLUSIONS: We propose that high levels of apo(a)/Lp(a), perhaps acting via a specific cell-surface binding domain, inhibit hepatic clearance of remnants, leading to high plasma levels of RLPs and markedly enhanced atherosclerosis. We speculate that the KIV(5-8) region of apo(a) competes with one or more receptors for remnant clearance in the liver and that this process may represent one mechanism accounting for increased atherosclerosis in humans with high secretion levels of apo(a).


Assuntos
Apolipoproteínas A/sangue , Apolipoproteínas A/genética , Aterosclerose/metabolismo , Colesterol/metabolismo , Quilomícrons/sangue , Lipoproteínas/metabolismo , Triglicerídeos/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Apolipoproteína B-100 , Apolipoproteína B-48 , Apolipoproteínas A/farmacologia , Apolipoproteínas B/sangue , Apolipoproteínas B/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , VLDL-Colesterol/sangue , Remanescentes de Quilomícrons , Feminino , Humanos , Kringles/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ratos , Triglicerídeos/sangue
9.
J Mol Med (Berl) ; 91(6): 749-58, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23361368

RESUMO

Inhibition of vascular endothelial growth factor increases response rates to chemotherapy and progression-free survival in glioblastoma. However, resistance invariably occurs, prompting the urgent need for identification of synergizing agents. One possible strategy is to understand tumor adaptation to microenvironmental changes induced by antiangiogenic drugs and test agents that exploit this process. We used an in vivo glioblastoma-derived xenograft model of tumor escape in presence of continuous treatment with bevacizumab. U87-MG or U118-MG cells were subcutaneously implanted into either BALB/c SCID or athymic nude mice. Bevacizumab was given by intraperitoneal injection every 3 days (2.5 mg/kg/dose) and/or dichloroacetate (DCA) was administered by oral gavage twice daily (50 mg/kg/dose) when tumor volumes reached 0.3 cm(3) and continued until tumors reached approximately 1.5-2.0 cm(3). Microarray analysis of resistant U87 tumors revealed coordinated changes at the level of metabolic genes, in particular, a widening gap between glycolysis and mitochondrial respiration. There was a highly significant difference between U87-MG-implanted athymic nude mice 1 week after drug treatment. By 2 weeks of treatment, bevacizumab and DCA together dramatically blocked tumor growth compared to either drug alone. Similar results were seen in athymic nude mice implanted with U118-MG cells. We demonstrate for the first time that reversal of the bevacizumab-induced shift in metabolism using DCA is detrimental to neoplastic growth in vivo. As DCA is viewed as a promising agent targeting tumor metabolism, our data establish the timely proof of concept that combining it with antiangiogenic therapy represents a potent antineoplastic strategy.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Ácido Dicloroacético/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Neoplasias/tratamento farmacológico , Animais , Bevacizumab , Linhagem Celular Tumoral , Quimioterapia Combinada , Feminino , Humanos , Hipóxia , Camundongos , Camundongos SCID , Neoplasias/patologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Cell Biol ; 189(6): 1027-38, 2010 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-20530211

RESUMO

Rapid plasma membrane resealing is essential for cellular survival. Earlier studies showed that plasma membrane repair requires Ca(2+)-dependent exocytosis of lysosomes and a rapid form of endocytosis that removes membrane lesions. However, the functional relationship between lysosomal exocytosis and the rapid endocytosis that follows membrane injury is unknown. In this study, we show that the lysosomal enzyme acid sphingomyelinase (ASM) is released extracellularly when cells are wounded in the presence of Ca(2+). ASM-deficient cells, including human cells from Niemann-Pick type A (NPA) patients, undergo lysosomal exocytosis after wounding but are defective in injury-dependent endocytosis and plasma membrane repair. Exogenously added recombinant human ASM restores endocytosis and resealing in ASM-depleted cells, suggesting that conversion of plasma membrane sphingomyelin to ceramide by this lysosomal enzyme promotes lesion internalization. These findings reveal a molecular mechanism for restoration of plasma membrane integrity through exocytosis of lysosomes and identify defective plasma membrane repair as a possible component of the severe pathology observed in NPA patients.


Assuntos
Membrana Celular/metabolismo , Endocitose/fisiologia , Exocitose/fisiologia , Esfingomielina Fosfodiesterase/metabolismo , Animais , Linhagem Celular , Membrana Celular/ultraestrutura , Ceramidas/metabolismo , Desipramina/metabolismo , Endossomos/metabolismo , Endossomos/ultraestrutura , Inibidores Enzimáticos/metabolismo , Inativação Gênica , Humanos , Lisossomos/enzimologia , Lisossomos/ultraestrutura , Doenças de Niemann-Pick/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Esfingomielina Fosfodiesterase/genética , Esfingomielinas/metabolismo
11.
PLoS One ; 5(4): e10345, 2010 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-20436681

RESUMO

BACKGROUND: Hypoxia in cancers results in the upregulation of hypoxia inducible factor 1 (HIF-1) and a microRNA, hsa-miR-210 (miR-210) which is associated with a poor prognosis. METHODS AND FINDINGS: In human cancer cell lines and tumours, we found that miR-210 targets the mitochondrial iron sulfur scaffold protein ISCU, required for assembly of iron-sulfur clusters, cofactors for key enzymes involved in the Krebs cycle, electron transport, and iron metabolism. Down regulation of ISCU was the major cause of induction of reactive oxygen species (ROS) in hypoxia. ISCU suppression reduced mitochondrial complex 1 activity and aconitase activity, caused a shift to glycolysis in normoxia and enhanced cell survival. Cancers with low ISCU had a worse prognosis. CONCLUSIONS: Induction of these major hallmarks of cancer show that a single microRNA, miR-210, mediates a new mechanism of adaptation to hypoxia, by regulating mitochondrial function via iron-sulfur cluster metabolism and free radical generation.


Assuntos
Ciclo do Ácido Cítrico , Radicais Livres/metabolismo , Hipóxia , Proteínas Ferro-Enxofre/metabolismo , MicroRNAs/fisiologia , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Linhagem Celular Tumoral , Complexo I de Transporte de Elétrons/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias/patologia , Prognóstico
12.
Curr Pharm Des ; 15(33): 3861-6, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19925433

RESUMO

Recent studies have established that the regulation of microRNAs (miRs) is a feature of the hypoxic response. In this review, we discuss the role of hypoxia-regulated miRs, with an emphasis on miR-210 and miR-373, and anticipate directions for clinical applications. The induction of miR-210 and miR-373 is dependent upon hypoxia inducible factor (HIF), and their up-regulation has been detected in a variety of solid tumors. Both miRs have been associated with adverse prognosis and metastatic potential. The increased expression of miR-210 is linked to an in vivo hypoxic signature. MiR-210 also participates in endothelial and neuronal cells' response to oxygen deprivation and may possess a role in the regulation of angiogenesis. A variety of miR-210 and miR-373 targets that may be relevant to hypoxia have been validated or proposed. Very recently, targets of these miRs that are implicated in DNA repair have been identified, thus establishing an additional link between the hypoxic tumor microenvironment and DNA damage. Extending beyond cancer biology, some of miR-210 targets are likely involved in the regulation of angiogenesis, and neuronal cell survival. Inactivation of miRs affected by hypoxia presents a promising therapeutic strategy in the case of difficult-to-treat cancers, as well as in other non-cancer-related diseases.


Assuntos
Sistemas de Liberação de Medicamentos , MicroRNAs/metabolismo , Neoplasias/genética , Animais , Hipóxia Celular/fisiologia , Reparo do DNA , Regulação da Expressão Gênica , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , MicroRNAs/genética , Neoplasias/tratamento farmacológico , Neoplasias/fisiopatologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/fisiopatologia
13.
J Biol Chem ; 280(31): 28299-305, 2005 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-15958390

RESUMO

Biological methylation reactions and homocysteine (Hcy) metabolism are intimately linked. In previous work, we have shown that phosphatidylethanolamine N-methyltransferase, an enzyme that methylates phosphatidylethanolamine to form phosphatidylcholine, plays a significant role in the regulation of plasma Hcy levels through an effect on methylation demand (Noga, A. A., Stead, L. M., Zhao, Y., Brosnan, M. E., Brosnan, J. T., and Vance, D. E. (2003) J. Biol. Chem. 278, 5952-5955). We have further investigated methylation demand and Hcy metabolism in liver-specific CTP:phosphocholine cytidylyltransferase-alpha (CTalpha) knockout mice, since flux through the phosphatidylethanolamine N-methyltransferase pathway is increased 2-fold to meet hepatic demand for phosphatidylcholine. Our data show that plasma Hcy is elevated by 20-40% in mice lacking hepatic CTalpha. CTalpha-deficient hepatocytes secrete 40% more Hcy into the medium than do control hepatocytes. Liver activity of betaine:homocysteine methyltransferase and methionine adenosyltransferase are elevated in the knockout mice as a mechanism for maintaining normal hepatic S-adenosylmethionine and S-adenosylhomocysteine levels. These data suggest that phospholipid methylation in the liver is a major consumer of AdoMet and a significant source of plasma Hcy.


Assuntos
Homocisteína/sangue , Fosfolipídeos/metabolismo , Animais , Betaína-Homocisteína S-Metiltransferase , Membrana Celular/metabolismo , Citidina Difosfato Colina/metabolismo , Citosol/metabolismo , Hepatócitos/metabolismo , Fígado/embriologia , Metionina Adenosiltransferase/metabolismo , Metilação , Metiltransferases/metabolismo , Camundongos , Camundongos Knockout , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/metabolismo
14.
Proc Natl Acad Sci U S A ; 99(9): 6280-5, 2002 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-11983917

RESUMO

Inflammatory cytokines have been linked to atherosclerosis by using cell culture models and acute inflammation in animals. The goal of this study was to examine lipoprotein levels and early atherosclerosis in chronic animal models of altered IL-1 physiology by using mice with deficient or excess IL-1 receptor antagonist (IL-1ra). IL-1ra knockout C57BL/6J mice fed a cholesterol/cholate diet for 3 mo had a 3-fold decrease in non-high-density lipoprotein cholesterol and a trend toward increased foam-cell lesion area compared to wild-type littermate controls. IL-1ra transgenic/low-density lipoprotein receptor (LDLR) knockout mice fed a cholesterol-saturated fat diet for 10 wk showed a 40% increase in non-high-density lipoprotein cholesterol, consistent with the IL-1ra knockout data, although there was no change in lesion size. When these IL1-ra overexpressing transgenic mice on the LDLR knockout background were fed a high-cholesterol/high-fat diet containing cholate, however, a statistically significant 40% decrease in lesion area was observed compared to LDLR knockout mice lacking the transgene. By immunohistochemistry, IL-1ra was present in C57BL/6J and LDLR knockout aortae, absent in IL-1ra knockout aortae, and present at high levels in LDLR knockout/IL-1ra transgene aortae. In summary, IL-1ra tended to increase plasma lipoprotein levels and, when fed a cholate-containing diet, decrease foam-cell lesion size. These data demonstrate that in selected models of murine atherosclerosis, chronic IL-1ra depletion or overexpression has potentially important effects on lipoprotein metabolism and foam-cell lesion development.


Assuntos
Colesterol/sangue , Células Espumosas/metabolismo , Mutação , Sialoglicoproteínas/genética , Sialoglicoproteínas/fisiologia , Animais , Colatos/farmacologia , Colesterol/farmacologia , Cruzamentos Genéticos , Endotélio Vascular/patologia , Genótipo , Imuno-Histoquímica , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/metabolismo , Lipoproteínas/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
15.
J Biol Chem ; 279(45): 47402-10, 2004 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-15331603

RESUMO

CTP:phosphocholine cytidylyltransferase (CT) is the key regulatory enzyme in the CDP-choline pathway for the biosynthesis of phosphatidylcholine. Hepatic cells express both an alpha and a beta2 isoform of CT and can also synthesize phosphatidylcholine via the sequential methylation of phosphatidylethanolamine catalyzed by phosphatidylethanolamine N-methyltransferase. To ascertain the functional importance of CTalpha, we created a mouse in which the hepatic CTalpha gene was specifically inactivated by the Cre/LoxP procedure. In CTalpha knockout mice, hepatic CT activity (due to residual CTbeta2 activity as well as activity in nonhepatic cells) was 15% of normal, whereas phosphatidylethanolamine N-methyltransferase activity was elevated 2-fold compared with controls. Lipid analyses of the liver indicated that female knockout mice had reduced phosphatidylcholine levels and accumulated triacylglycerols. The plasma phosphatidylcholine concentration was reduced in the CTalpha knockout (independent of gender), as were levels of high density lipoproteins (cholesterol and apoAI) and very low density lipoproteins (triacylglycerols and apoB100). Experiments in which mice were injected with Triton WR1339 indicated that apoB secretion was decreased in hepatic-specific CTalpha knockout mice compared with controls. These results suggest an important role for hepatic CTalpha in regulating both hepatic and systemic lipid and lipoprotein metabolism.


Assuntos
Colina-Fosfato Citidililtransferase/genética , Lipoproteínas HDL/sangue , Lipoproteínas LDL/sangue , Animais , Apolipoproteínas B/metabolismo , Bile/metabolismo , Colesterol/metabolismo , Ésteres do Colesterol/metabolismo , Colina-Fosfato Citidililtransferase/química , Colina-Fosfato Citidililtransferase/fisiologia , Detergentes/farmacologia , Feminino , Deleção de Genes , Homozigoto , Immunoblotting , Metabolismo dos Lipídeos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fosfatidilcolinas/química , Polietilenoglicóis/farmacologia , Isoformas de Proteínas , Fatores Sexuais , Fatores de Tempo , Triglicerídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA