Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
FEBS Open Bio ; 14(2): 300-308, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38105414

RESUMO

Expression of α-smooth muscle actin (αSMA) is constitutive in vascular smooth muscle cells, but is induced in nonmuscle cells such as hepatic stellate cells (HSCs). HSCs play important roles in both physiological homeostasis and pathological response. HSC activation is characterized by αSMA expression, which is regulated by the TGFß-induced Smad pathway. Recently, protein kinase C (PKC) was identified to regulate αSMA expression. Diacylglycerol kinase (DGK) metabolizes a second-messenger DG, thereby controlling components of DG-mediated signaling, such as PKC. In the present study we aimed to investigate the putative role of DGKα in αSMA expression. Use of a cellular model indicated that the DGK inhibitor R59949 promotes αSMA expression and PKCδ phosphorylation. It also facilitates Smad2 phosphorylation after 30 min of TGFß stimulation. Furthermore, immunocytochemical analysis revealed that DGK inhibitor pretreatment without TGFß stimulation engenders αSMA expression in a granular pattern, whereas DGK inhibitor pretreatment plus TGFß stimulation significantly induces αSMA incorporation in stress fibers. Through animal model experiments, we observed that DGKα-knockout mice exhibit increased expression of αSMA in the liver after 48 h of carbon tetrachloride injection, together with enhanced phosphorylation levels of Smad2 and PKCδ. Together, these findings suggest that DGKα negatively regulates αSMA expression by acting on the Smad and PKCδ signaling pathways, which differentially regulate stress fiber incorporation and protein expression of αSMA, respectively.


Assuntos
Actinas , Fígado , Animais , Camundongos , Actinas/metabolismo , Fígado/metabolismo , Músculo Liso/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta , Diacilglicerol Quinase
3.
Adv Biol Regul ; 75: 100659, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31607681

RESUMO

Adipose tissue is a central site for energy storage in the form of triglyceride (TG). Under excess energy conditions, TG is synthesized by acylation of diacylglycerol (DG), whereas TG is broken down into DG and free fatty acid, which provide energy for mitochondrial lipid oxidation when needed. In this regard, DG is not merely an intermediate metabolite for TG metabolism; it also serves as a signaling molecule. DG kinase (DGK) phosphorylates DG to produce phosphatidic acid (PA). Consequently, DGK plays a pivotal role in the control of lipid metabolism and signal transduction pathway. Recently, a report has described that DGKε-knockout (KO) mice show expansion of epididymal white adipose tissue (WAT) together with the impairment of glucose clearance after short-term (40 days) high fat diet (HFD) feeding, an early presymptomatic phase of obesity in wild-type animals. Nevertheless, no report describes an investigation of their phenotype under long-term HFD feeding conditions. Remarkably, results obtained during long-term HFD feeding show that WAT mass is decreased significantly and that the blood glucose profile in response to glucose challenge is improved in DGKε-KO mice compared with wild-type, which contrast sharply against the phenotype shown for short-term HFD feeding. Morphological examination reveals that cyclooxygenase-2 (COX-2) expression and clusters of uncoupling protein 1 (UCP1)-positive multilocular brown-like ("beige") adipocyte are induced in DGKε-deficient WAT after long-term HFD feeding, suggesting that beige adipocytes facilitate energy expenditure during prolonged HFD feeding. Administration of celecoxib, a selective inhibitor of COX-2, abolishes the appearance of UCP1-positive beige adipocytes in DGKε-KO mice. These findings suggest that DGKε deficiency promotes visceral WAT remodeling in a COX-2-dependent manner under long-term HFD feeding conditions.


Assuntos
Adipogenia , Tecido Adiposo Bege/enzimologia , Ciclo-Oxigenase 2/metabolismo , Diacilglicerol Quinase/deficiência , Gorduras na Dieta/farmacologia , Gordura Intra-Abdominal/enzimologia , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Ciclo-Oxigenase 2/genética , Diacilglicerol Quinase/metabolismo , Camundongos , Camundongos Knockout
4.
FASEB J ; 32(8): 4121-4131, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29509511

RESUMO

Lipid metabolism is closely involved with signal transduction and energy homeostasis. Excess calorie intake causes abnormal lipid metabolism, promoting obesity and insulin resistance. Diacylglycerol (DG) represents not only a lipidic second messenger but also an intermediate metabolite for triglyceride metabolism in the endoplasmic reticulum (ER). However, it remains undetermined how the roles of DG in signaling and energy homeostasis is regulated within the cell. Of DG kinases (DGKs), which are enzymes that phosphorylate DG, DGKε resides in the ER. This study examined how DGKε is implicated in signal transduction and lipid homeostasis. DGKε-deficient mice were fed a high-fat diet (HFD) for 40 d. We observed that DGKε deficiency promotes fat accumulation in adipocytes and subsequently promotes insulin resistance in mice fed an HFD. This abnormal fat metabolism is mediated by down-regulation of lipolytic activities, such as adipose triglyceride lipase and hormone-sensitive lipase. In addition, activation of DG-sensitive PKC leads to insulin resistance in adipose tissue, which may be caused by delayed metabolism of DG. Our data suggest that DGKε links the second messenger signaling system to energy homeostasis in adipocytes and that its deficiency results in abnormal lipid metabolism such as obesity and insulin resistance.-Nakano, T., Seino, K., Wakabayashi, I., Stafforini, D. M., Topham, M. K., Goto, K. Deletion of diacylglycerol kinase ε confers susceptibility to obesity via reduced lipolytic activity in murine adipocytes.


Assuntos
Adipócitos/metabolismo , Diacilglicerol Quinase/metabolismo , Obesidade/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Regulação para Baixo/fisiologia , Homeostase/fisiologia , Resistência à Insulina/fisiologia , Lipase/metabolismo , Metabolismo dos Lipídeos/fisiologia , Camundongos , Transdução de Sinais/fisiologia
5.
Cell Tissue Res ; 368(3): 441-458, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28191598

RESUMO

Following activation of Gq protein-coupled receptors, phospholipase C yields a pair of second messengers: diacylglycerol (DG) and inositol 1,4,5-trisphosphate. Diacylglycerol kinase (DGK) phosphorylates DG to produce phosphatidic acid, another second messenger. Of the DGK family, DGKε is the only DGK isoform that exhibits substrate specificity for DG with an arachidonoyl acyl chain at the sn-2 position. Recently, we demonstrated that hydrophobic residues in the N-terminus of DGKε play an important role in targeting the endoplasmic reticulum in transfected cells. However, its cellular expression and subcellular localization in the brain remain elusive. In the present study, we investigate this issue using specific DGKε antibody. DGKε was richly expressed in principal neurons of higher brain regions, including pyramidal cells in the hippocampus and neocortex, medium spiny neurons in the striatum and Purkinje cells in the cerebellum. In Purkinje cells, DGKε was localized to the subsurface cisterns and colocalized with inositol 1,4,5-trisphosphate receptor-1 in dendrites and axons. In dendrites of Purkinje cells, DGKε was also distributed in close apposition to DG lipase-α, which catalyzes arachidonoyl-DG to produce 2-arachidonoyl glycerol, a major endocannabinoid in the brain. Behaviorally, DGKε-knockout mice exhibited hyper-locomotive activities and impaired motor coordination and learning. These findings suggest that DGKε plays an important role in neuronal and brain functions through its distinct neuronal expression and subcellular localization and also through coordinated arrangement with other molecules involving the phosphoinositide signaling pathway.


Assuntos
Cerebelo/enzimologia , Diacilglicerol Quinase/metabolismo , Células de Purkinje/enzimologia , Animais , Encéfalo/enzimologia , Cerebelo/citologia , Cerebelo/ultraestrutura , Diacilglicerol Quinase/genética , Células HeLa , Humanos , Immunoblotting , Imuno-Histoquímica , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Aprendizagem , Locomoção , Camundongos , Camundongos Knockout , Células PC12 , Fosfatidilinositóis/metabolismo , Desempenho Psicomotor , Células de Purkinje/ultraestrutura , Ratos , Ratos Wistar , Sistemas do Segundo Mensageiro , Distribuição Tecidual
6.
J Cell Physiol ; 232(3): 617-624, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27312515

RESUMO

Bone homeostasis is maintained by a balance between resorption of the bone matrix and its replacement by new bone. Osteoclasts play a crucially important role in bone metabolism. They are responsible for bone resorption under pathophysiological conditions. Differentiation of these cells, which are derived from bone marrow cells, depends on receptor activator of NF-κB ligand (RANKL). RANKL-induced osteoclastogenesis is regulated by the phosphoinositide (PI) signaling pathway, in which diacylglycerol (DG) serves as a second messenger in signal transduction. In this study, we examined the functional implications of DG kinase (DGK), an enzyme family responsible for DG metabolism, for osteoclast differentiation and activity. Of DGKs, DGKζ is most abundantly expressed in osteoclast precursors such as bone marrow-derived monocytes/macrophages. During osteoclast differentiation from precursor cells, DGKζ is downregulated at the protein level. In this regard, we found that DGKζ deletion enhances osteoclast differentiation and bone resorption activity under inflammatory conditions in an animal model of osteolysis. Furthermore, DGKζ deficiency upregulates RANKL expression in response to TNFα stimulation. Collectively, results suggest that DGKζ is silent under normal conditions, but it serves as a negative regulator in osteoclast function under inflammatory conditions. Downregulation of DGKζ might be one factor predisposing a person to osteolytic bone destruction in pathological conditions. J. Cell. Physiol. 232: 617-624, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Reabsorção Óssea/enzimologia , Reabsorção Óssea/patologia , Diferenciação Celular , Diacilglicerol Quinase/metabolismo , Regulação para Baixo , Inflamação/patologia , Osteoclastos/patologia , Animais , Biomarcadores/metabolismo , Densidade Óssea/efeitos dos fármacos , Reabsorção Óssea/complicações , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Fibroblastos/metabolismo , Inflamação/complicações , Inflamação/enzimologia , Isoenzimas/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Knockout , Osteoclastos/efeitos dos fármacos , Osteólise/complicações , Osteólise/enzimologia , Osteólise/patologia , Ligante RANK/genética , Ligante RANK/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Tempo , Tomografia Computadorizada por Raios X , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima/efeitos dos fármacos
7.
Biochim Biophys Acta ; 1861(12 Pt A): 1993-1999, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27713003

RESUMO

The incorporation of glycerol into lipid was measured using SV40 transformed mouse embryo fibroblasts (MEFs) from either wild-type (WT) mice or from mice in which the epsilon isoform of diacylglycerol kinase (DGKε) was knocked out (DGKε-/-). We present an explanation for our finding that DGKε-/- MEFs exhibited greater uptake of 3H-glycerol into the cell and a greater incorporation into lipids compared with their WT counterparts, with no change in the relative amounts of various lipids between the DGKε-/- and WT MEFs. Glycerol kinase is more highly expressed in the DGKε-/- cells than in their WT counterparts. In addition, the activity of glycerol kinase is greater in the DGKε-/- cells than in their WT counterparts. Other substrates that enter the cell independent of glycerol kinase, such as pyruvate or acetate, are incorporated into lipid to the same extent between DGKε-/- and WT cell lines. We also show that expression of p53, a transcription factor that increases the synthesis of glycerol kinase, is increased in DGKε-/- MEFs in comparison to WT cells. We conclude that the increased incorporation of glycerol into lipids in DGKε-/- cells is a consequence of up-regulation of glycerol kinase and not a result of an increase in the rate of lipid synthesis. Furthermore, increased expression of the pro-survival gene, p53, in cells knocked out for DGKε suggests that cells over-expressing DGKε would have a greater propensity to become tumorigenic.


Assuntos
Diacilglicerol Quinase/metabolismo , Fibroblastos/metabolismo , Glicerol Quinase/metabolismo , Glicerol/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular , Lipídeos/fisiologia , Lipogênese/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição/metabolismo , Regulação para Cima/fisiologia
8.
JAMA ; 315(12): 1266-75, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27002448

RESUMO

IMPORTANCE: Patients with familial adenomatous polyposis (FAP) are at markedly increased risk for duodenal polyps and cancer. Surgical and endoscopic management of duodenal neoplasia is difficult and chemoprevention has not been successful. OBJECTIVE: To evaluate the effect of a combination of sulindac and erlotinib on duodenal adenoma regression in patients with FAP. DESIGN, SETTING, AND PARTICIPANTS: Double-blind, randomized, placebo-controlled trial, enrolling 92 participants with FAP, conducted from July 2010 through June 2014 at Huntsman Cancer Institute in Salt Lake City, Utah. INTERVENTIONS: Participants with FAP were randomized to sulindac (150 mg) twice daily and erlotinib (75 mg) daily (n = 46) vs placebo (n = 46) for 6 months. MAIN OUTCOMES AND MEASURES: The total number and diameter of polyps in the proximal duodenum were mapped at baseline and 6 months. The primary outcome was change in total polyp burden at 6 months. Polyp burden was calculated as the sum of the diameters of polyps. The secondary outcomes were change in total duodenal polyp count, change in duodenal polyp burden or count stratified by genotype and initial polyp burden, and percentage of change from baseline in duodenal polyp burden. RESULTS: Ninety-two participants (mean age, 41 years [range, 24-55]; women, 56 [61%]) were randomized when the trial was stopped by the external data and safety monitoring board because the second preplanned interim analysis met the prespecified stopping rule for superiority. Grade 1 and 2 adverse events were more common in the sulindac-erlotinib group, with an acne-like rash observed in 87% of participants receiving treatment and 20% of participants receiving placebo (P < .001). Only 2 participants experienced grade 3 adverse events. [table: see text]. CONCLUSIONS AND RELEVANCE: Among participants with FAP, the use of sulindac and erlotinib compared with placebo resulted in a lower duodenal polyp burden after 6 months. Adverse events may limit the use of these medications at the doses used in this study. Further research is necessary to evaluate these preliminary findings in a larger study population with longer follow-up to determine whether the observed effects will result in improved clinical outcomes. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT 01187901.


Assuntos
Polipose Adenomatosa do Colo/tratamento farmacológico , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Duodenais/tratamento farmacológico , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/patologia , Adulto , Antineoplásicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias Duodenais/genética , Neoplasias Duodenais/patologia , Cloridrato de Erlotinib/administração & dosagem , Cloridrato de Erlotinib/efeitos adversos , Feminino , Genes APC , Humanos , Masculino , Pessoa de Meia-Idade , Sulindaco/administração & dosagem , Sulindaco/efeitos adversos
9.
Am J Physiol Renal Physiol ; 310(9): F895-908, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26887830

RESUMO

Thrombotic microangiopathy (TMA) is a disorder characterized by microvascular occlusion that can lead to thrombocytopenia, hemolytic anemia, and glomerular damage. Complement activation is the central event in most cases of TMA. Primary forms of TMA are caused by mutations in genes encoding components of the complement or regulators of the complement cascade. Recently, we and others have described a genetic form of TMA caused by mutations in the gene diacylglycerol kinase-ε (DGKE) that encodes the lipid kinase DGKε (Lemaire M, Fremeaux-Bacchi V, Schaefer F, Choi MR, Tang WH, Le Quintrec M, Fakhouri F, Taque S, Nobili F, Martinez F, Ji WZ, Overton JD, Mane SM, Nurnberg G, Altmuller J, Thiele H, Morin D, Deschenes G, Baudouin V, Llanas B, Collard L, Majid MA, Simkova E, Nurnberg P, Rioux-Leclerc N, Moeckel GW, Gubler MC, Hwa J, Loirat C, Lifton RP. Nat Genet 45: 531-536, 2013; Ozaltin F, Li BH, Rauhauser A, An SW, Soylemezoglu O, Gonul II, Taskiran EZ, Ibsirlioglu T, Korkmaz E, Bilginer Y, Duzova A, Ozen S, Topaloglu R, Besbas N, Ashraf S, Du Y, Liang CY, Chen P, Lu DM, Vadnagara K, Arbuckle S, Lewis D, Wakeland B, Quigg RJ, Ransom RF, Wakeland EK, Topham MK, Bazan NG, Mohan C, Hildebrandt F, Bakkaloglu A, Huang CL, Attanasio M. J Am Soc Nephrol 24: 377-384, 2013). DGKε is unrelated to the complement pathway, which suggests that unidentified pathogenic mechanisms independent of complement dysregulation may result in TMA. Studying Dgke knockout mice may help to understand the pathogenesis of this disease, but no glomerular phenotype has been described in these animals so far. Here we report that Dgke null mice present subclinical microscopic anomalies of the glomerular endothelium and basal membrane that worsen with age and develop glomerular capillary occlusion when exposed to nephrotoxic serum. We found that induction of cyclooxygenase-2 and of the proangiogenic prostaglandin E2 are impaired in Dgke null kidneys and are associated with reduced expression of the antithrombotic cell adhesion molecule platelet endothelial cell adhesion molecule-1/CD31 in the glomerular endothelium. Notably, prostaglandin E2 supplementation was able to rescue motility defects of Dgke knockdown cells in vitro and to restore angiogenesis in a test in vivo. Our results unveil an unexpected role of Dgke in the induction of cyclooxygenase-2 and in the regulation of glomerular prostanoids synthesis under stress.


Assuntos
Ciclo-Oxigenase 2/biossíntese , Diacilglicerol Quinase/genética , Dinoprostona/biossíntese , Endotélio/patologia , Glomerulonefrite/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Envelhecimento/patologia , Animais , Movimento Celular , Glomerulonefrite/enzimologia , Glomerulonefrite/metabolismo , Testes de Função Renal , Glomérulos Renais/enzimologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica , Cicatrização
10.
Biochim Biophys Acta ; 1853(2): 361-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25450975

RESUMO

The transcription factor NF-κB family serves as a key component of many pathophysiological events such as innate and adaptive immune response, inflammation, apoptosis, and oncogenesis. Various cell signals trigger activation of the regulatory mechanisms of NF-κB, resulting in its nuclear translocation and transcriptional initiation. The diacylglycerol kinase (DGK) family, a lipid second messenger-metabolizing enzyme in phosphoinositide signaling, is shown to regulate widely various cellular processes. Results of recent studies suggest that one family member, DGKζ, is closely involved in immune and inflammatory responses. Nevertheless, little is known about the regulatory mechanism of DGKζ on NF-κB pathway in cytokine-induced inflammatory signaling. This study shows that siRNA-mediated DGKζ knockdown in HeLa cells facilitates degradation of IκB, followed by nuclear translocation of NF-κB p65 subunit. In addition, DGKζ-deficient MEFs show upregulation of p65 subunit phosphorylation at Serine 468 and 536 and its interaction with CBP transcriptional coactivator upon TNF-α stimulation. These modifications of p65 subunit might engender enhanced NF-κB transcriptional reporter assay of DGKζ knockdown cells. These findings provide further insight into the regulatory mechanisms of cytokine-induced NF-κB activation.


Assuntos
Citocinas/farmacologia , Diacilglicerol Quinase/metabolismo , Regulação para Baixo/efeitos dos fármacos , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Proteína de Ligação a CREB/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Proteínas I-kappa B/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Interferência de RNA/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
11.
IUBMB Life ; 66(10): 694-703, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25355554

RESUMO

We previously found that conditional deletion of integrin ß1 in intestinal epithelium of mice caused early postnatal lethality and intestinal phenotypic changes including excessive proliferation and defective differentiation of intestinal epithelium due to loss of Hedgehog expression. Here, we link these defects to the Hedgehog (Hh) signaling pathway and show that loss of integrin ß1 leads to excessive phosphorylation of MEK-1 and increased expression of ErbB receptors, including the epidermal growth factor receptor (EGFR). We show that increased EGFR signaling attenuates Hh abundance and that an EGFR inhibitor rescues conditional ß1 integrin null pups from postnatal lethality. These studies link the loss of Hh expression in the intestinal epithelium of integrin ß1-deficient mice to excessive EGFR/MAPK signaling, and identify a unique mechanism for crosstalk between stromal and epithelial signaling pathways that is critical for intestinal epithelial differentiation and function.


Assuntos
Receptores ErbB/metabolismo , Regulação da Expressão Gênica , Proteínas Hedgehog/genética , Integrina beta1/fisiologia , Mucosa Intestinal/metabolismo , Proteínas dos Microfilamentos/fisiologia , Animais , Western Blotting , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Receptores ErbB/genética , Feminino , Proteínas Hedgehog/metabolismo , Técnicas Imunoenzimáticas , Integrases/metabolismo , Mucosa Intestinal/citologia , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
12.
Biochim Biophys Acta ; 1842(10): 1440-50, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25048194

RESUMO

The endoplasmic reticulum (ER), comprised of an interconnected membrane network, is a site of phospholipid and protein synthesis. The diacylglycerol kinase (DGK) enzyme family catalyzes phosphorylation of diacylglycerol to phosphatidic acid. Both of these lipids are known not only to serve as second messengers but also to represent intermediate precursors of lipids of various kinds. The DGK family is targeted to distinct subcellular sites in cDNA-transfected and native cells. Of DGKs, DGKε localizes primarily to the ER, suggesting that this isozyme plays a role in this organelle. Using experiments with various deletion and substitution mutants, this study examined the molecular mechanism of how DGKε is targeted to the ER. Results demonstrate that the N-terminal hydrophobic sequence 20-40 plays a necessary role in targeting of DGKε to the ER. This hydrophobic amino acid segment is predicted to adopt an α-helix structure, in which Leu22, L25, and L29 are present in mutual proximity, forming a hydrophobic patch. When these hydrophobic Leu residues were replaced with hydrophilic amino acid Gln, the mutant fragment designated DGKε (20-40/L22Q,L25Q,L29Q) exhibits diffuse distribution in the cytoplasm. Moreover, full-length DGKε containing these substitutions, DGKε (L22Q,L25Q,L29Q), is shown to distribute diffusely in the cytoplasm. These results indicate that the N-terminal hydrophobic residues play a key role in DGKε targeting to the ER membrane. Functionally, knockdown or deletion of DGKε affects the unfolding protein response pathways, thereby rendering the cells susceptible to apoptosis, to some degree, under ER stress conditions.

13.
Adv Biol Regul ; 54: 242-53, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24119575

RESUMO

Eukaryotic cells have evolved to possess a distinct subcellular compartment, the nucleus, separated from the cytoplasm in a manner that allows the precise operation of the chromatin, thereby permitting controlled access to the regulatory elements in the DNA for transcription and replication. In the cytoplasm, genetic information contained in the DNA sequence is translated into proteins, including enzymes that catalyze various reactions, such as metabolic processes, energy control, and responses to changing environments. One mechanism that regulates these events involves phosphoinositide turnover signaling, which generates a lipid second messenger, diacylglycerol (DG). Since DG acts as a potent activator of several signaling molecules, it should be tightly regulated to keep cellular responsiveness within a physiological range. DG kinase (DGK) metabolizes DG by phosphorylating it to generate phosphatidic acid, thus serving as a critical regulator of DG signaling. Phosphoinositide turnover is employed differentially in the nucleus and the cytoplasm. A member of the DGK family, DGKζ, localizes to the nucleus in various cell types and is considered to regulate nuclear DG signaling. Recent studies have provided evidence that DGKζ shuttles between the nucleus and the cytoplasm in neurons under pathophysiological conditions. Transport of a signal regulator between the nucleus and the cytoplasm should be a critical function for maintaining basic processes in the nucleus, such as cell cycle regulation and gene expression, and to ensure communication between nuclear processes and cytoplasmic functions. In this review, a series of studies on nucleocytoplasmic translocation of DGKζ have been summarized, and the functional implications of this phenomenon in postmitotic neurons and cancer cells under stress conditions are discussed.


Assuntos
Núcleo Celular/enzimologia , Citoplasma/enzimologia , Diacilglicerol Quinase/metabolismo , Neoplasias/enzimologia , Neoplasias/fisiopatologia , Animais , Núcleo Celular/genética , Citoplasma/genética , Diacilglicerol Quinase/genética , Diglicerídeos/metabolismo , Humanos , Neoplasias/genética , Transporte Proteico , Estresse Fisiológico
14.
Biochemistry ; 52(44): 7766-76, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24090246

RESUMO

We have studied the relationship between diacylglycerol kinase delta (DGKδ) and lipogenesis. There is a marked increase in the expression of DGKδ during the differentiation of 3T3-L1 cells to adipocytes, as well as in the synthesis of neutral and polar lipids. When 3T3-L1 undifferentiated fibroblasts are transfected to express DGKδ, there is increased triglyceride synthesis without differentiation to adipocytes. Hence, expression of DGKδ promotes lipogenesis. Lipid synthesis is decreased in DGKδ knockout mouse embryo fibroblasts, especially for lipids with shorter acyl chains and limited unsaturation. This reduction occurs for both neutral and polar lipids. These findings suggest reduced de novo lipid synthesis. This is confirmed by measuring the incorporation of glycerol into polar and neutral lipids, which is higher in the wild type cells than in the DGKδ knockouts. In comparison, there was no change in lipid synthesis in DGKε knockout mouse embryo fibroblasts. We also demonstrate that the DGKδ knockout cells had a lower expression of acetyl-CoA carboxylase and fatty acid synthase as well as a lower degree of activation by phosphorylation of ATP citrate lyase. These three enzymes are involved in the synthesis of long chain fatty acids. Our results demonstrate that DGKδ markedly increases lipid synthesis, at least in part as a result of promoting the de novo synthesis of fatty acids.


Assuntos
Adipócitos/enzimologia , Diacilglicerol Quinase/metabolismo , Lipídeos/biossíntese , Lipogênese , Regulação para Cima , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Diacilglicerol Quinase/genética , Ácidos Graxos/biossíntese , Fibroblastos/citologia , Fibroblastos/metabolismo , Lipídeos/química , Masculino , Camundongos , Camundongos Knockout , Triglicerídeos/biossíntese
15.
J Immunol ; 191(4): 1907-15, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23851686

RESUMO

IFN-ε is a unique type I IFN whose constitutive expression in lung, brain, small intestine, and reproductive tissues is only partially understood. Our previous observation that posttranscriptional events participate in the regulation of IFN-ε mRNA expression led us to investigate whether the 5' and/or 3' untranslated regions (UTR) have regulatory functions. Surprisingly, we found that full-length IFN-ε 5'UTR markedly suppressed mRNA expression under basal conditions. Analysis of the secondary structure of this region predicted formation of two stable stem-loop structures, loops 1 and 2. Studies using luciferase constructs harboring various stretches of IFN-ε 5'UTR and mutant constructs in which the conformation of loop structures was disrupted showed that loop 1 is essential for regulation of mRNA expression. Incubation of HeLa cell extracts with agarose-bound RNAs harboring IFN-ε loop structures identified importin 9 (IPO9), a molecular transporter and chaperone, as a candidate that associates with these regions of the 5'UTR. IPO9 overexpression decreased, and IPO9 silencing increased basal IFN-ε expression. Our studies uncover a previously undescribed function for IPO9 as a specific, and negative, posttranscriptional regulator of IFN-ε expression, and they identify key roles for IFN-ε stem-loop structure 1 in this process. IPO9-mediated effects on 5'UTRs appear to extend to additional mRNAs, including hypoxia-inducible factor-1α, that can form specific loop structures.


Assuntos
Regiões 5' não Traduzidas/genética , Regulação da Expressão Gênica/fisiologia , Interferons/genética , Sequências Repetidas Invertidas/genética , Carioferinas/fisiologia , Interferência de RNA , Animais , Sequência de Bases , Sequência Consenso , Regulação para Baixo , Feminino , Genes Reporter , Células HeLa , Humanos , Interferons/biossíntese , Carioferinas/genética , Mamíferos/genética , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Estabilidade de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Neoplasias do Colo do Útero/patologia
16.
J Cell Sci ; 126(Pt 13): 2785-97, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23606744

RESUMO

The transcription factor p53 plays a crucial role in coordinating the cellular response to various stresses. Therefore, p53 protein levels and activity need to be kept under tight control. We report here that diacylglycerol kinase ζ (DGKζ) binds to p53 and modulates its function both in the cytoplasm and nucleus. DGKζ, a member of the DGK family that metabolizes a lipid second messenger diacylglycerol, localizes primarily to the nucleus in various cell types. Recently, reports have described that excitotoxic stress induces DGKζ nucleocytoplasmic translocation in hippocampal neurons. In the study reported here we found that cytoplasmic DGKζ attenuates p53-mediated cytotoxicity against doxorubicin-induced DNA damage by facilitating cytoplasmic anchoring and degradation of p53 through a ubiquitin-proteasome system. Concomitantly, decreased levels of nuclear DGKζ engender downregulation of p53 transcriptional activity. Consistent with these in vitro cellular experiments, DGKζ-deficient brain exhibits high levels of p53 protein after kainate-induced seizures and even under normal conditions. These findings provide novel insights into the regulation of p53 function and suggest that DGKζ serves as a sentinel to control p53 function both during normal homeostasis and in stress responses.


Assuntos
Citoplasma/metabolismo , Diacilglicerol Quinase/metabolismo , Diglicerídeos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Sobrevivência Celular , Citoplasma/efeitos dos fármacos , Citoplasma/genética , Dano ao DNA , Diacilglicerol Quinase/genética , Doxorrubicina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Injeções Intraperitoneais , Ácido Caínico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Transdução de Sinais , Transcrição Gênica , Proteína Supressora de Tumor p53/genética , Ubiquitina/genética , Ubiquitina/metabolismo
17.
J Am Soc Nephrol ; 24(3): 377-84, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23274426

RESUMO

Renal microangiopathies and membranoproliferative GN (MPGN) can manifest similar clinical presentations and histology, suggesting the possibility of a common underlying mechanism in some cases. Here, we performed homozygosity mapping and whole exome sequencing in a Turkish consanguineous family and identified DGKE gene variants as the cause of a membranoproliferative-like glomerular microangiopathy. Furthermore, we identified two additional DGKE variants in a cohort of 142 unrelated patients diagnosed with membranoproliferative GN. This gene encodes the diacylglycerol kinase DGKε, which is an intracellular lipid kinase that phosphorylates diacylglycerol to phosphatidic acid. Immunofluorescence confocal microscopy demonstrated that mouse and rat Dgkε colocalizes with the podocyte marker WT1 but not with the endothelial marker CD31. Patch-clamp experiments in human embryonic kidney (HEK293) cells showed that DGKε variants affect the intracellular concentration of diacylglycerol. Taken together, these results not only identify a genetic cause of a glomerular microangiopathy but also suggest that the phosphatidylinositol cycle, which requires DGKE, is critical to the normal function of podocytes.


Assuntos
Diacilglicerol Quinase/genética , Glomerulonefrite Membranoproliferativa/enzimologia , Glomerulonefrite Membranoproliferativa/genética , Nefropatias/enzimologia , Nefropatias/genética , Mutação , Sequência de Aminoácidos , Animais , Sequência de Bases , Estudos de Coortes , Consanguinidade , DNA/genética , Diacilglicerol Quinase/metabolismo , Diagnóstico Diferencial , Diglicerídeos/metabolismo , Feminino , Variação Genética , Glomerulonefrite Membranoproliferativa/patologia , Células HEK293 , Humanos , Nefropatias/patologia , Glomérulos Renais/enzimologia , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem , Podócitos/metabolismo , Polimorfismo de Nucleotídeo Único , Ratos , Homologia de Sequência de Aminoácidos , Turquia
18.
Cancer Res ; 73(9): 2806-16, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23361301

RESUMO

Platelet-activating factor (PAF) is a naturally occurring phospholipid that mediates diverse effects such as physiological and pathological inflammation, immunosuppression, and cancer. Several lines of evidence support both positive and negative roles for PAF in carcinogenesis. PAF stimulates cell growth, oncogenic transformation, and metastasis, but can also limit proliferation and induce apoptosis. The biological context and microenvironment seem to define whether PAF has pro- or anticarcinogenic effects. To investigate the role of exacerbated PAF signaling in colon cancer, we conducted cell-based and in vivo studies using genetically engineered mice lacking expression of phospholipase A2 group 7 (PLA2G7), an enzyme that specifically metabolizes PAF and structurally related glycerophospholipids. Absence of Pla2g7 robustly decreased intestinal polyposis and colon tumor formation in Apc(Min)(/+) mice, suggesting an antitumorigenic role for PAF in settings characterized by aberrant function of the tumor suppressor Adenomatous polyposis coli (Apc). In colonic epithelial cells, exposure to a PAF analog led to dephosphorylation of Akt at serine-473 and induction of apoptosis. The mechanism of this response involved formation of a complex between ß-arrestin 1 and the Akt phosphatase PHLPP2, and activation of the intrinsic pathway of apoptosis. Our results suggest that strategies based on inhibiting PLA2G7 activity or increasing PAF-mediated signaling hold promise for the treatment of intestinal malignancies that harbor mutations in APC.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Neoplasias do Colo/patologia , Polipose Intestinal/metabolismo , Fosfolipases A2/genética , Fosfolipases A2/fisiologia , 1-Alquil-2-acetilglicerofosfocolina Esterase , Alelos , Animais , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Células Epiteliais/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Imuno-Histoquímica/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Fosforilação , Transdução de Sinais
19.
J Biol Chem ; 288(3): 1439-47, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23184957

RESUMO

Discovering proteins that modulate Akt signaling has become a critical task, given the oncogenic role of Akt in a wide variety of cancers. We have discovered a novel diacylglycerol signaling pathway that promotes dephosphorylation of Akt. This pathway is regulated by diacylglycerol kinase δ (DGKδ). In DGKδ-deficient cells, we found reduced Akt phosphorylation downstream of three receptor tyrosine kinases. Phosphorylation upstream of Akt was not affected. Our data indicate that PKCα, which is excessively active in DGKδ-deficient cells, promotes dephosphorylation of Akt through pleckstrin homology domain leucine-rich repeats protein phosphatase (PHLPP) 2. Depletion of either PKCα or PHLPP2 rescued Akt phosphorylation in DGKδ-deficient cells. In contrast, depletion of PHLPP1, another Akt phosphatase, failed to rescue Akt phosphorylation. Other PHLPP substrates were not affected by DGKδ deficiency, suggesting mechanisms allowing specific modulation of Akt dephosphorylation. We found that ß-arrestin 1 acted as a scaffold for PHLPP2 and Akt1, providing a mechanism for specificity. Because of its ability to reduce Akt phosphorylation, we tested whether depletion of DGKδ could attenuate tumorigenic properties of cultured cells and found that DGKδ deficiency reduced cell proliferation and migration and enhanced apoptosis. We have, thus, discovered a novel pathway in which diacylglycerol signaling negatively regulates Akt activity. Our collective data indicate that DGKδ is a pertinent cancer target, and our studies could lay the groundwork for development of novel cancer therapeutics.


Assuntos
Diacilglicerol Quinase/metabolismo , Diglicerídeos/metabolismo , Fosfoproteínas Fosfatases/deficiência , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Arrestinas/genética , Arrestinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica , Diacilglicerol Quinase/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Vetores Genéticos , Humanos , Lentivirus/genética , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases/genética , Fosforilação , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/genética , Especificidade por Substrato , beta-Arrestina 1 , beta-Arrestinas
20.
Chem Phys Lipids ; 166: 26-30, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23261795

RESUMO

The diacylglycerol kinase from E. coli transfers some of the γ-phosphate of ATP to water as well as to diacylglycerol. We also demonstrate that glycerol can act as an acceptor for the phosphate of ATP. We have compared this behavior with that of the only mammalian isoform of diacylglycerol kinase that exhibits acyl chain specificity, i.e. DGKɛ. The purpose of the study was to determine if differences in the competition between ATPase activity and lipid phosphorylation could contribute to the observed acyl chain specificity with different diacylglycerols. Neither with the highly specific substrate of DGKɛ, 1-stearoyl-2-arachidonoyl glycerol, nor with a less specific substrate, 1-stearoyl-2-linoleoyl glycerol, is there any evidence for ATP hydrolysis accompanying substrate phosphorylation. Thus, at least for this isoform of diacylglycerol kinase, water does not compete with diacylglycerol as an acceptor of the γ-phosphate of ATP. The results demonstrate that the substrate specificity of mammalian DGKɛ is not a consequence of different degrees of ATP hydrolysis in the presence of different species of diacylglycerol.


Assuntos
Trifosfato de Adenosina/metabolismo , Diacilglicerol Quinase/metabolismo , Diglicerídeos/metabolismo , Animais , Linhagem Celular , Escherichia coli/enzimologia , Humanos , Hidrólise , Ressonância Magnética Nuclear Biomolecular , Fosforilação , Isoformas de Proteínas/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...