Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(16)2021 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-34445455

RESUMEN

Glycerol is used in many skin care products because it improves skin function. Anecdotal reports by patients on the National Psoriasis Foundation website also suggest that glycerol may be helpful for the treatment of psoriasis, although to date no experimental data confirm this idea. Glycerol entry into epidermal keratinocytes is facilitated by aquaglyceroporins like aquaporin-3 (AQP3), and its conversion to phosphatidylglycerol, a lipid messenger that promotes keratinocyte differentiation, requires the lipid-metabolizing enzyme phospholipase-D2 (PLD2). To evaluate whether glycerol inhibits inflammation and psoriasiform lesion development in the imiquimod (IMQ)-induced mouse model of psoriasis, glycerol's effect on psoriasiform skin lesions was determined in IMQ-treated wild-type and PLD2 knockout mice, with glycerol provided either in drinking water or applied topically. Psoriasis area and severity index, ear thickness and ear biopsy weight, epidermal thickness, and inflammatory markers were quantified. Topical and oral glycerol ameliorated psoriasiform lesion development in wild-type mice. Topical glycerol appeared to act as an emollient to induce beneficial effects, since even in PLD2 knockout mice topical glycerol application improved skin lesions. In contrast, the beneficial effects of oral glycerol required PLD2, with no improvement in psoriasiform lesions observed in PLD2 knockout mice. Our findings suggest that the ability of oral glycerol to improve psoriasiform lesions requires its PLD2-mediated conversion to phosphatidylglycerol, consistent with our previous report that phosphatidylglycerol itself improves psoriasiform lesions in this model. Our data also support anecdotal evidence that glycerol can ameliorate psoriasis symptoms and therefore might be a useful therapy alone or in conjunction with other treatments.


Asunto(s)
Glicerol/farmacología , Imiquimod/efectos adversos , Psoriasis/tratamiento farmacológico , Piel/metabolismo , Animales , Acuaporina 3/genética , Acuaporina 3/metabolismo , Modelos Animales de Enfermedad , Humanos , Imiquimod/farmacología , Ratones , Ratones Noqueados , Fosfolipasa D/deficiencia , Fosfolipasa D/metabolismo , Psoriasis/inducido químicamente , Psoriasis/genética , Psoriasis/metabolismo
2.
FASEB J ; 33(3): 3167-3179, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30399323

RESUMEN

Oleoylethanolamide (OEA), a fatty acid ethanolamide (FAE), is a lipid mediator that controls food intake and lipid metabolism. Accumulating data imply the importance of intestinal OEA in controlling satiety in addition to gastrointestinal peptide hormones. Although the biochemical pathway of FAE production has been illustrated, the enzymes responsible for the cleavage of OEA from its precursor N-acyl-phosphatidylethanolamine (NAPE) must be identified among reported candidates in the gut. In this study, we assessed the involvement of NAPE-specific phospholipase D (NAPE-PLD), which can directly release FAEs from NAPE, in intestinal OEA synthesis and lipid metabolism. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPER-associated protein 9 (Cas9)-mediated deletion of the NAPE-PLD gene in intestinal epithelial-like Caco-2 cells reduced OEA levels, regardless of their differentiation states. Transcriptome analysis revealed that deletion of NAPE-PLD activates a transcriptional program for nutrient transportation, including lipids and lipoproteins, and inactivates cell-cycle or mitosis-related genes in Caco-2 cells. In addition, the basolateral secretion of lipoproteins was increased in NAPE-PLD-deleted cells although lipoprotein size was not affected. By contrast, cellular lipid levels were reduced in NAPE-PLD-deleted cells. Overall, these results indicate that NAPE-PLD plays important roles in OEA synthesis and fat absorption by regulating lipoprotein production in the intestinal epithelial cells.-Igarashi, M., Watanabe, K., Tsuduki, T., Kimura, I., Kubota, N. NAPE-PLD controls OEA synthesis and fat absorption by regulating lipoprotein synthesis in an in vitro model of intestinal epithelial cells.


Asunto(s)
Grasas de la Dieta/metabolismo , Endocannabinoides/biosíntesis , Mucosa Intestinal/metabolismo , Ácidos Oléicos/biosíntesis , Fosfolipasa D/metabolismo , Antígenos CD36/metabolismo , Células CACO-2 , Diferenciación Celular , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Absorción Intestinal/genética , Absorción Intestinal/fisiología , Mucosa Intestinal/citología , Metabolismo de los Lípidos , Lipoproteínas/biosíntesis , Modelos Biológicos , Fosfolipasa D/deficiencia , Fosfolipasa D/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Nature ; 505(7484): 550-554, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24336208

RESUMEN

Genome-wide association studies (GWAS) have identified several risk variants for late-onset Alzheimer's disease (LOAD). These common variants have replicable but small effects on LOAD risk and generally do not have obvious functional effects. Low-frequency coding variants, not detected by GWAS, are predicted to include functional variants with larger effects on risk. To identify low-frequency coding variants with large effects on LOAD risk, we carried out whole-exome sequencing (WES) in 14 large LOAD families and follow-up analyses of the candidate variants in several large LOAD case-control data sets. A rare variant in PLD3 (phospholipase D3; Val232Met) segregated with disease status in two independent families and doubled risk for Alzheimer's disease in seven independent case-control series with a total of more than 11,000 cases and controls of European descent. Gene-based burden analyses in 4,387 cases and controls of European descent and 302 African American cases and controls, with complete sequence data for PLD3, reveal that several variants in this gene increase risk for Alzheimer's disease in both populations. PLD3 is highly expressed in brain regions that are vulnerable to Alzheimer's disease pathology, including hippocampus and cortex, and is expressed at significantly lower levels in neurons from Alzheimer's disease brains compared to control brains. Overexpression of PLD3 leads to a significant decrease in intracellular amyloid-ß precursor protein (APP) and extracellular Aß42 and Aß40 (the 42- and 40-residue isoforms of the amyloid-ß peptide), and knockdown of PLD3 leads to a significant increase in extracellular Aß42 and Aß40. Together, our genetic and functional data indicate that carriers of PLD3 coding variants have a twofold increased risk for LOAD and that PLD3 influences APP processing. This study provides an example of how densely affected families may help to identify rare variants with large effects on risk for disease or other complex traits.


Asunto(s)
Enfermedad de Alzheimer/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Fosfolipasa D/genética , Negro o Afroamericano/genética , Edad de Inicio , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Estudios de Casos y Controles , Europa (Continente)/etnología , Exoma/genética , Femenino , Humanos , Masculino , Fragmentos de Péptidos/metabolismo , Fosfolipasa D/deficiencia , Fosfolipasa D/metabolismo , Procesamiento Proteico-Postraduccional/genética , Proteolisis
4.
Int J Mol Sci ; 21(9)2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-32370031

RESUMEN

BACKGROUND: Phospholipase (PL)D1 is crucial for integrin αIIbß3 activation of platelets in arterial thrombosis and TNF-α-mediated inflammation and TGF-ß-mediated collagen scar formation after myocardial infarction (MI) in mice. Enzymatic activity of PLD is not responsible for PLD-mediated TNF-α signaling and myocardial healing. The impact of PLD2 in ischemia reperfusion injury is unknown. METHODS: PLD2-deficient mice underwent myocardial ischemia and reperfusion (I/R). RESULTS: Enhanced integrin αIIbß3 activation of platelets resulted in elevated interleukin (IL)-6 release from endothelial cells in vitro and enhanced IL-6 plasma levels after MI in PLD2-deficient mice. This was accompanied by enhanced migration of inflammatory cells into the infarct border zone and reduced TGF-ß plasma levels after 72 h that might account for enhanced inflammation in PLD2-deficient mice. In contrast to PLD1, TNF-α signaling, infarct size and cardiac function 24 h after I/R were not altered when PLD2 was deleted. Furthermore, TGF-ß plasma levels, scar formation and heart function were comparable between PLD2-deficient and control mice 21 days post MI. CONCLUSIONS: The present study contributes to our understanding about the role of PLD isoforms and altered platelet signaling in the process of myocardial I/R injury.


Asunto(s)
Plaquetas/metabolismo , Integrinas/metabolismo , Infarto del Miocardio/complicaciones , Miocarditis/etiología , Miocarditis/metabolismo , Fosfolipasa D/deficiencia , Animales , Biomarcadores , Supervivencia Celular , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Células Endoteliales/metabolismo , Expresión Génica , Integrinas/química , Masculino , Ratones , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocarditis/patología
5.
J Immunol ; 195(9): 4492-502, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26392467

RESUMEN

Phospholipase D (PLD) proteins are enzymes that catalyze the hydrolysis of phosphatidylcholine to generate an important signaling lipid, phosphatidic acid. Phosphatidic acid is a putative second messenger implicated in the regulation of vesicular trafficking and cytoskeletal reorganization. Previous studies using inhibitors and overexpression of PLD proteins indicate that PLD1 and PLD2 play positive roles in FcεRI-mediated signaling and mast cell function. We used mice deficient in PLD1, PLD2, or both to study the function of these enzymes in mast cells. In contrast to published studies, we found that PLD1 deficiency impaired FcεRI-mediated mast cell degranulation; however, PLD2 deficiency enhanced it. Biochemical analysis showed that PLD deficiency affected activation of the PI3K pathway and RhoA. Furthermore, our data indicated that, although PLD1 deficiency impaired F-actin disassembly, PLD2 deficiency enhanced microtubule formation. Together, our results suggested that PLD1 and PLD2, two proteins that catalyze the same enzymatic reaction, regulate different steps in mast cell degranulation.


Asunto(s)
Mastocitos/inmunología , Fosfolipasa D/inmunología , Receptores de IgE/inmunología , Transducción de Señal/inmunología , Actinas/inmunología , Actinas/metabolismo , Animales , Western Blotting , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Degranulación de la Célula/inmunología , Células Cultivadas , Citoesqueleto/inmunología , Citoesqueleto/metabolismo , Mastocitos/metabolismo , Mastocitos/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Fosfatidilinositol 3-Quinasas/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfolipasa D/deficiencia , Fosfolipasa D/genética , Receptores de IgE/metabolismo , Proteína de Unión al GTP rhoA/inmunología , Proteína de Unión al GTP rhoA/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 34(8): 1697-703, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24947526

RESUMEN

OBJECTIVE: Aberrant regulation of the proliferation, survival, and migration of endothelial cells (ECs) is closely related to the abnormal angiogenesis that occurs in hypoxia-induced pathological situations, such as cancer and vascular retinopathy. Hypoxic conditions and the subsequent upregulation of hypoxia-inducible factor-1α and target genes are important for the angiogenic functions of ECs. Phospholipase D2 (PLD2) is a crucial signaling mediator that stimulates the production of the second messenger phosphatidic acid. PLD2 is involved in various cellular functions; however, its specific roles in ECs under hypoxia and in vivo angiogenesis remain unclear. In the present study, we investigated the potential roles of PLD2 in ECs under hypoxia and in hypoxia-induced pathological angiogenesis in vivo. APPROACH AND RESULTS: Pld2 knockout ECs exhibited decreased hypoxia-induced cellular responses in survival, migration, and thus vessel sprouting. Analysis of hypoxia-induced gene expression revealed that PLD2 deficiency disrupted the upregulation of hypoxia-inducible factor-1α target genes, including VEGF, PFKFB3, HMOX-1, and NTRK2. Consistent with this, PLD2 contributed to hypoxia-induced hypoxia-inducible factor-1α expression at the translational level. The roles of PLD2 in hypoxia-induced in vivo pathological angiogenesis were assessed using oxygen-induced retinopathy and tumor implantation models in endothelial-specific Pld2 knockout mice. Pld2 endothelial-specific knockout retinae showed decreased neovascular tuft formation, despite a larger avascular region. Tumor growth and tumor blood vessel formation were also reduced in Pld2 endothelial-specific knockout mice. CONCLUSIONS: Our findings demonstrate a novel role for endothelial PLD2 in the survival and migration of ECs under hypoxia via the expression of hypoxia-inducible factor-1α and in pathological retinal angiogenesis and tumor angiogenesis in vivo.


Asunto(s)
Carcinoma Pulmonar de Lewis/irrigación sanguínea , Células Endoteliales/enzimología , Hipoxia/complicaciones , Neovascularización Patológica , Fosfolipasa D/deficiencia , Neovascularización Retiniana/enzimología , Vasos Retinianos/enzimología , Animales , Animales Recién Nacidos , Hipoxia de la Célula , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfolipasa D/genética , Interferencia de ARN , Neovascularización Retiniana/etiología , Neovascularización Retiniana/genética , Neovascularización Retiniana/patología , Vasos Retinianos/patología , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección
7.
Arterioscler Thromb Vasc Biol ; 33(9): 2212-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23868933

RESUMEN

OBJECTIVE: We recently showed that mice lacking the lipid signaling enzyme phospholipase (PL) D1 or both PLD isoforms (PLD1 and PLD2) were protected from pathological thrombus formation and ischemic stroke, whereas hemostasis was not impaired in these animals. We sought to assess whether pharmacological inhibition of PLD activity affects hemostasis, thrombosis, and thrombo-inflammatory brain infarction in mice. APPROACH AND RESULTS: Treatment of platelets with the reversible, small molecule PLD inhibitor, 5-fluoro-2-indolyl des-chlorohalopemide (FIPI), led to a specific blockade of PLD activity that was associated with reduced α-granule release and integrin activation. Mice that received FIPI at a dose of 3 mg/kg displayed reduced occlusive thrombus formation upon chemical injury of carotid arteries or mesenterial arterioles. Similarly, FIPI-treated mice had smaller infarct sizes and significantly better motor and neurological function 24 hours after transient middle cerebral artery occlusion. This protective effect was not associated with major intracerebral hemorrhage or prolonged tail bleeding times. CONCLUSIONS: These results provide the first evidence that pharmacological PLD inhibition might provide a safe therapeutic strategy to prevent arterial thrombosis and ischemic stroke.


Asunto(s)
Plaquetas/efectos de los fármacos , Enfermedades de las Arterias Carótidas/prevención & control , Domperidona/análogos & derivados , Inhibidores Enzimáticos/farmacología , Fibrinolíticos/farmacología , Indoles/farmacología , Infarto de la Arteria Cerebral Media/prevención & control , Fosfolipasa D/antagonistas & inhibidores , Trombosis/prevención & control , Animales , Plaquetas/enzimología , Enfermedades de las Arterias Carótidas/sangre , Enfermedades de las Arterias Carótidas/enzimología , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/fisiopatología , Modelos Animales de Enfermedad , Domperidona/farmacología , Relación Dosis-Respuesta a Droga , Hemostasis/efectos de los fármacos , Infarto de la Arteria Cerebral Media/sangre , Infarto de la Arteria Cerebral Media/enzimología , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/fisiopatología , Integrinas/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfolipasa D/deficiencia , Fosfolipasa D/genética , Recuperación de la Función , Trombosis/sangre , Trombosis/enzimología , Trombosis/genética , Trombosis/fisiopatología , Factores de Tiempo
8.
J Lipid Res ; 54(11): 3151-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24018423

RESUMEN

Lipid aldehydes including isolevuglandins (IsoLGs) and 4-hydroxynonenal modify phosphatidylethanolamine (PE) to form proinflammatory and cytotoxic adducts. Therefore, cells may have evolved mechanisms to degrade and prevent accumulation of these potentially harmful compounds. To test if cells could degrade isolevuglandin-modified phosphatidylethanolamine (IsoLG-PE), we generated IsoLG-PE in human embryonic kidney 293 (HEK293) cells and human umbilical cord endothelial cells and measured its stability over time. We found that IsoLG-PE levels decreased more than 75% after 6 h, suggesting that IsoLG-PE was indeed degraded. Because N-acyl phosphatidylethanolamine-hydrolyzing phospholipase D (NAPE-PLD) has been described as a key enzyme in the hydrolysis of N-acyl phosphatidylethanoamine (NAPE) and both NAPE and IsoLG-PE have large aliphatic headgroups, we considered the possibility that this enzyme might also hydrolyze IsoLG-PE. We found that knockdown of NAPE-PLD expression using small interfering RNA (siRNA) significantly increased the persistence of IsoLG-PE in HEK293 cells. IsoLG-PE competed with NAPE for hydrolysis by recombinant mouse NAPE-PLD, with the catalytic efficiency (V(max)/K(m)) for hydrolysis of IsoLG-PE being 30% of that for hydrolysis of NAPE. LC-MS/MS analysis confirmed that recombinant NAPE-PLD hydrolyzed IsoLG-PE to IsoLG-ethanolamine. These results demonstrate that NAPE-PLD contributes to the degradation of IsoLG-PE and suggest that a major physiological role of NAPE-PLD may be to degrade aldehyde-modified PE, thereby preventing the accumulation of these harmful compounds.


Asunto(s)
Aldehídos/metabolismo , Fosfatidiletanolaminas/metabolismo , Fosfolipasa D/metabolismo , Animales , Silenciador del Gen , Células HEK293 , Humanos , Hidrólisis , Ratones , Fosfolipasa D/deficiencia , Fosfolipasa D/genética
9.
J Lipid Res ; 54(2): 379-85, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23213138

RESUMEN

Functional defects in ATPase class I type 8B membrane 1 (ATP8B1 or familial intrahepatic cholestasis 1, FIC1) lead to cholestasis by mechanism(s) that are not fully understood. One proposed pathophysiology involves aberrant signaling to the bile acid sensor, the farnesoid X receptor (FXR), via protein kinase C ζ (PKCζ). The following cell line-based studies investigated whether phospholipase D2 may transduce a signal from FIC1 to FXR. PLD2 gain of function led to activation of the bile salt export pump (BSEP) promoter, a well-characterized FXR response. BSEP activation by PLD2 could be blocked by abrogating either PKCζ or FXR signaling. PLD2 loss of function led to a reduction in BSEP promoter activity. In addition, a variety of proteins that are activated by FXR, including BSEP, were reduced in HepG2 cells treated with PLD2 siRNA. Similar effects were observed in freshly isolated human hepatocytes. Activation of BSEP by FIC1 gain of function was blocked when PLD2 but not PLD1 was silenced. Overexpression of wild-type but not Byler mutant FIC1 led to an increase in membrane associated PLD activity. An intermediate level of activation of PLD activity was induced when a benign recurrent intrahepatic cholestasis FIC1 mutant construct was expressed. These studies show that FIC1 signals to FXR via a signaling pathway including PLD2 and PKCζ.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Fosfolipasa D/metabolismo , Transducción de Señal , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/genética , Animales , Células CHO , Cricetinae , Cricetulus , Silenciador del Gen , Células Hep G2 , Humanos , Fosfolipasa D/deficiencia , Fosfolipasa D/genética , Regiones Promotoras Genéticas/genética , Proteína Quinasa C/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo
10.
J Cell Sci ; 124(Pt 12): 1973-83, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21610093

RESUMEN

The signalling lipid phosphatidic acid (PA) is generated by the hydrolysis of phosphatidylcholine (PC), which is catalysed by phospholipase D (PLD) enzymes. Neutrophils, important cells of the innate immune system, maintain the body's defence against infection. Previous studies have implicated PLD-generated PA in neutrophil function; these have relied heavily on the use of primary alcohols to act as inhibitors of PA production. The recent development of isoform-selective small molecule inhibitors and the generation of a knockout mouse model provide us with accurate tools to study the role of PLDs in neutrophil responses. We show that PLD1 is a regulator of phorbol-ester-, chemoattractant, adhesion-dependent and Fcγ-receptor-stimulated production of reactive oxygen species (ROS) in neutrophils. Significantly we found that this role of PLD is isoform specific: the absence of PLD2 does not negatively affect these processes. Contrary to expectation, other functions required for an efficient immune response operate effectively in Pld2-deficient neutrophils or when both isoforms are inhibited pharmacologically. We conclude that although PLD1 does have important regulatory roles in neutrophils, the field has been confused by the use of primary alcohols; now that gold standard Pld-knockout mouse models are available, previous work might need to be reassessed.


Asunto(s)
Neutrófilos/metabolismo , Fosfolipasa D/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de IgG/metabolismo , Animales , Adhesión Celular/fisiología , Ratones , Ratones Noqueados , Ésteres del Forbol , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/deficiencia , Fosfolipasa D/genética , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología
11.
J Neurosci ; 30(49): 16419-28, 2010 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-21147981

RESUMEN

Growing evidence implicates aberrant lipid signaling in Alzheimer's disease (AD). While phospholipases A2 and C have been recently shown to mediate key actions of amyloid ß-peptide (Aß) through a dysregulation of arachidonic acid and phosphatidylinositol-4,5-bisphosphate metabolism, respectively, the role of phospholipase D (PLD) has so far remained elusive. PLD produces phosphatidic acid (PA), a bioactive lipid involved in multiple aspects of cell physiology, including signaling and membrane trafficking processes. Here we show that oligomeric Aß enhances PLD activity in cultured neurons and that this stimulatory effect does not occur upon ablation of PLD2 via gene targeting. Aß fails to suppress long-term potentiation in PLD2-deficient hippocampal slices, suggesting that PLD2 is required for the synaptotoxic action of this peptide. In vivo PLD activity, as assessed by detection of phosphatidylethanol levels using mass spectrometry (MS) following ethanol injection, is also increased in the brain of a transgenic mouse model of AD (SwAPP). Furthermore, Pld2 ablation rescues memory deficits and confers synaptic protection in SwAPP mice despite a significant Aß load. MS-based lipid analysis of Pld2 mutant brains in the presence or absence of the SwAPP transgene unmasks striking crosstalks between different PA species. This lipid analysis shows an exquisite acyl chain specificity and plasticity in the perturbation of PA metabolism. Collectively, our results point to specific molecular species of PA as key modulators of AD pathogenesis and identify PLD2 as a novel potential target for therapeutics.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Fosfolipasa D/deficiencia , Sinapsis/genética , Factores de Edad , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Conducta Animal , Línea Celular Transformada , Trastornos del Conocimiento/genética , Condicionamiento Psicológico/fisiología , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Estrenos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Miedo/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , Humanos , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Espectrometría de Masas/métodos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Células PC12/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Fosfolipasa D/genética , Pirrolidinonas/farmacología , Ratas , Factores de Tiempo
12.
Am J Physiol Cell Physiol ; 298(1): C163-70, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19864325

RESUMEN

Lysophosphatidic acid (LPA), is a lipid mediator that binds to G-protein coupled receptors. Epidermal growth factor (EGF), a polypeptide growth factor, binds to the EGF receptor (EGFR), a receptor tyrosine kinase. Both LPA and EGF induce responses in tumor cells that include proliferation, migration, metastasis, and induction of angiogenesis. LPA has the potential to act as an autocrine/paracrine factor and can transactivate the EGFR. This study explores the role of phospholipase D2 (PLD2) activation in LPA production, as well as cross-talk between EGF and LPA receptors. We demonstrate that EGF and LPA both stimulate production of LPA by OVCAR3 and SKOV3 human ovarian cancer cell lines. PD158780, an EGFR-selective tyrosine kinase inhibitor, blocks LPA production in response to both EGF and LPA in OVCAR3 and SKOV3 cells. Pertussis toxin, an inhibitor of LPA receptor signaling, inhibits LPA production in response to both EGF and LPA. Similar results were observed for the LPA receptor antagonist, Ki16425. Overexpression of PLD2 increases LPA production, while knockdown of PLD2 blocks EGF-induced LPA production. A phospholipase A2 (PLA2) inhibitor also blocks LPA- and EGF-induced LPA production. These results indicate that EGF stimulates LPA production in a manner that requires PLD2, and suggest that cross-talk can occur bidirectionally between EGF and LPA receptors.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Lisofosfolípidos/biosíntesis , Neoplasias Ováricas/metabolismo , Toxina del Pertussis/farmacología , Fosfolipasa D/metabolismo , Receptores del Ácido Lisofosfatídico/fisiología , Línea Celular Tumoral , Receptores ErbB/efectos de los fármacos , Receptores ErbB/fisiología , Femenino , Vectores Genéticos , Humanos , Fosfolipasa D/deficiencia , Fosfolipasa D/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Receptor Cross-Talk/fisiología , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores del Ácido Lisofosfatídico/efectos de los fármacos , Activación Transcripcional , Transfección
13.
Infect Immun ; 78(5): 1952-62, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20194595

RESUMEN

Acinetobacter baumannii is an emerging bacterial pathogen of considerable health care concern. Nonetheless, relatively little is known about the organism's virulence factors or their regulatory networks. Septicemia and ventilator-associated pneumonia are two of the more severe forms of A. baumannii disease. To identify virulence factors that may contribute to these disease processes, genetically diverse A. baumannii clinical isolates were evaluated for the ability to proliferate in human serum. A transposon mutant library was created in a strain background that propagated well in serum and screened for members with decreased serum growth. The results revealed that disruption of A. baumannii phospholipase D (PLD) caused a reduction in the organism's ability to thrive in serum, a deficiency in epithelial cell invasion, and diminished pathogenesis in a murine model of pneumonia. Collectively, these results suggest that PLD is an A. baumannii virulence factor.


Asunto(s)
Infecciones por Acinetobacter/patología , Acinetobacter baumannii/patogenicidad , Proteínas Bacterianas/genética , Fosfolipasa D/deficiencia , Factores de Virulencia/deficiencia , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/crecimiento & desarrollo , Secuencia de Aminoácidos , Estructuras Animales/microbiología , Animales , Recuento de Colonia Microbiana , Elementos Transponibles de ADN , Células Epiteliales/microbiología , Histocitoquímica , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía , Datos de Secuencia Molecular , Mutagénesis Insercional , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Suero/microbiología , Virulencia
14.
Prog Lipid Res ; 78: 101018, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31830503

RESUMEN

Despite being discovered over 60 years ago, the precise role of phospholipase D (PLD) is still being elucidated. PLD enzymes catalyze the hydrolysis of the phosphodiester bond of glycerophospholipids producing phosphatidic acid and the free headgroup. PLD family members are found in organisms ranging from viruses, and bacteria to plants, and mammals. They display a range of substrate specificities, are regulated by a diverse range of molecules, and have been implicated in a broad range of cellular processes including receptor signaling, cytoskeletal regulation and membrane trafficking. Recent technological advances including: the development of PLD knockout mice, isoform-specific antibodies, and specific inhibitors are finally permitting a thorough analysis of the in vivo role of mammalian PLDs. These studies are facilitating increased recognition of PLD's role in disease states including cancers and Alzheimer's disease, offering potential as a target for therapeutic intervention.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/metabolismo , Animales , Humanos , Fosfolipasa D/deficiencia
15.
Neuropharmacology ; 54(1): 1-7, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17631919

RESUMEN

Endocannabinoids, including anandamide (arachidonoyl ethanolamide) have been implicated in the regulation of a growing number of physiological and pathological processes. Anandamide can be generated from its membrane phospholipid precursor N-arachidonoyl phosphatidylethanolamine (NAPE) through hydrolysis by a phospholipase D (NAPE-PLD). Recent evidence indicates, however, the existence of two additional, parallel pathways. One involves the sequential deacylation of NAPE by alpha,beta-hydrolase 4 (Abhd4) and the subsequent cleavage of glycerophosphate to yield anandamide, and the other one proceeds through phospholipase C-mediated hydrolysis of NAPE to yield phosphoanandamide, which is then dephosphorylated by phosphatases, including the tyrosine phosphatase PTPN22 and the inositol 5' phosphatase SHIP1. Conversion of synthetic NAPE to AEA by brain homogenates from wild-type and NAPE-PLD(-/-) mice can proceed through both the PLC/phosphatase and Abdh4 pathways, with the former being dominant at shorter (<10 min) and the latter at longer (60 min) incubations. In macrophages, the endotoxin-induced synthesis of anandamide proceeds uniquely through the phospholipase C/phosphatase pathway.


Asunto(s)
Ácidos Araquidónicos/biosíntesis , Redes y Vías Metabólicas/fisiología , Animales , Línea Celular Transformada , Cromatografía Líquida de Alta Presión , Cromatografía en Capa Delgada , Interacciones Farmacológicas , Endocannabinoides , Glicerofosfatos/metabolismo , Hidrolasas/metabolismo , Hidrólisis/efectos de los fármacos , Inositol Polifosfato 5-Fosfatasas , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Ratones Noqueados , Neomicina/farmacología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Fosfolipasa D/deficiencia , Monoéster Fosfórico Hidrolasas/deficiencia , Alcamidas Poliinsaturadas , Inhibidores de la Síntesis de la Proteína/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/deficiencia , ARN Interferente Pequeño/metabolismo , Transfección/métodos , Fosfolipasas de Tipo C/metabolismo
16.
Sci Rep ; 8(1): 10006, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29968773

RESUMEN

Sepsis is a systemic inflammatory disorder with organ dysfunction and represents the leading cause of mortality in non-coronary intensive care units. A key player in septic shock is Tumor Necrosis Factor-alpha (TNF-α). Phospholipase (PL)D1 is involved in the regulation of TNF-α upon ischemia/reperfusion injury in mice. In this study we analyzed the impact of PLD1 in the regulation of TNF-α, inflammation and organ damage in experimental sepsis. PLD1 deficiency increased survival of mice and decreased vital organ damage after LPS injections. Decreased TNF-α plasma levels and reduced migration of leukocytes and platelets into lungs was associated with reduced apoptosis in lung and liver tissue of PLD1 deficient mice. PLD1 deficient platelets contribute to preserved outcome after LPS-induced sepsis because platelets exhibit an integrin activation defect suggesting reduced platelet activation in PLD1 deficient mice. Furthermore, reduced thrombin generation of PLD1 deficient platelets might be responsible for reduced fibrin formation in lungs suggesting reduced disseminated intravascular coagulation (DIC). The analysis of Pld1fl/fl-PF4-Cre mice revealed that migration of neutrophils and cell apoptosis in septic animals is not due to platelet-mediated processes. The present study has identified PLD1 as a regulator of innate immunity that may be a new target to modulate sepsis.


Asunto(s)
Lipopolisacáridos/toxicidad , Fosfolipasa D/metabolismo , Choque Séptico/patología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/inmunología , Plaquetas/metabolismo , Movimiento Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Fibrina/metabolismo , Inmunidad Innata/inmunología , Inflamación/patología , Leucocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Fosfolipasa D/deficiencia , Fosfolipasa D/genética , Activación Plaquetaria/genética
17.
J Biochem ; 162(6): 449-458, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28992041

RESUMEN

N-acylethanolamines (NAEs), a class of lipid mediators, are produced from N-acyl-phosphatidylethanolamine (NAPE) by several pathways, including the direct release by NAPE-specific phospholipase D (NAPE-PLD) or the multistep pathway via sn-glycero-3-phospho-N-acylethanolamine (Gp-NAE). Using liquid chromatography-tandem mass spectrometry, we compared peripheral tissue levels of NAPE, Gp-NAE and NAE in NAPE-PLD-deficient (NAPE-PLD-/-) and wild type (WT) mice. NAPE-PLD was suggested to play a major role in the NAPE degradation in heart, kidney, and liver, but not in jejunum, because the NAPE levels except jejunum were significantly higher in NAPE-PLD-/- mice than in WT mice. The deletion of NAPE-PLD failed to alter the NAE levels of these tissues, suggesting its limited role in the NAE production. The enzyme assays with tissue homogenates confirmed the presence of NAPE-PLD-independent pathways in these peripheral tissues. Gp-NAE species having an acyl moiety with 22 carbons and 6 double bonds was enriched in these peripheral tissues. As for sn-2 acyl species of NAPE, 18:2-acyl-containing NAPE species were predominant over 18:1-containing species in heart, liver, and jejunum. Our results show that both molecular species composition of NAPE, NAE and Gp-NAE and their dependencies on Napepld are different among the peripheral tissues, suggesting that each tissue has distinct metabolic pathways and these NAE-containing lipids play tissue-specific roles.


Asunto(s)
Fosfatidiletanolaminas/química , Fosfolipasa D/metabolismo , Animales , Encéfalo , Etanolaminas/química , Etanolaminas/metabolismo , Corazón , Yeyuno/química , Riñón/química , Lípidos/análisis , Hígado/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Fosfatidiletanolaminas/metabolismo , Fosfolipasa D/deficiencia
18.
Cell Signal ; 38: 171-181, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28711718

RESUMEN

Glycoprotein (GP)Ib is not only required for stable thrombus formation but for platelet-mediated inflammatory responses. Phospholipase (PL)D1 is essential for GPIb-dependent aggregate formation under high shear conditions while nothing is known about PLD1-induced regulation of GPIb in platelet-mediated inflammation and the underlying mechanisms. This study aimed to investigate the relevance of PLD1 for platelet-mediated endothelial and leukocyte recruitment and activation in vitro and in vivo. Pld1-/- platelets showed strongly reduced adhesion to TNFα stimulated endothelial cells (ECs) under high shear conditions ex vivo. Normal cytoskeletal reorganization of Pld1-/- platelets but reduced integrin activation after adhesion to inflamed ECs confirmed that defective integrin activation is responsible for reduced platelet adhesion to ECs. This, together with significantly reduced CD40L expression on platelets led to reduced chemotactic and adhesive properties of ECs in vitro. Under flow conditions, recruitment of leukocytes to collagen-adherent platelets was reduced. Under inflammatory conditions in vivo, reduced platelet and leukocyte recruitment and arrest to the injured carotid artery was observed in Pld1-/- mice. In a second in vivo model of venous thrombosis, platelet adhesion to activated endothelial cells was reduced while leukocyte recruitment was attenuated in PLD1 deficient mice. Mechanistically, PLD1 modulates PLCγ2 phosphorylation and integrin activation via Src kinases without affecting vWF binding to GPIb. Thus, PLD1 is important for GPIb-induced inflammatory processes of platelets and might be a promising target to reduce platelet-mediated inflammation.


Asunto(s)
Plaquetas/enzimología , Plaquetas/patología , Inflamación/enzimología , Inflamación/patología , Fosfolipasa D/metabolismo , Animales , Adhesión Celular , Quimiotaxis , Citoesqueleto/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Leucocitos/patología , Ratones , Fosfolipasa C gamma/metabolismo , Fosfolipasa D/deficiencia , Fosforilación , Complejo GPIb-IX de Glicoproteína Plaquetaria , Resistencia al Corte , Transducción de Señal , Familia-src Quinasas/metabolismo
19.
Sci Rep ; 7(1): 9112, 2017 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-28831159

RESUMEN

The Phospholipase D (PLD) superfamily is linked to neurological disease, cancer, and fertility, and a recent report correlated a potential loss-of-function PLD2 polymorphism with hypotension. Surprisingly, PLD2 -/- mice exhibit elevated blood pressure accompanied by associated changes in cardiac performance and molecular markers, but do not have findings consistent with the metabolic syndrome. Instead, expression of endothelial nitric oxide synthase (eNOS), which generates the potent vasodilator nitric oxide (NO), is decreased. An eNOS inhibitor phenocopied PLD2 loss and had no further effect on PLD2 -/- mice, confirming the functional relationship. Using a human endothelial cell line, PLD2 loss of function was shown to lower intracellular free cholesterol, causing upregulation of HMG Co-A reductase, the rate-limiting enzyme in cholesterol synthesis. HMG Co-A reductase negatively regulates eNOS, and the PLD2-deficiency phenotype of decreased eNOS expression and activity could be rescued by cholesterol supplementation and HMG Co-A reductase inhibition. Together, these findings identify a novel pathway through which the lipid signaling enzyme PLD2 regulates blood pressure, creating implications for on-going therapeutic development of PLD small molecule inhibitors. Finally, we show that the human PLD2 polymorphism does not trigger eNOS loss, but rather creates another effect, suggesting altered functioning for the allele.


Asunto(s)
Presión Sanguínea/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfolipasa D/deficiencia , Transducción de Señal , Animales , Colesterol/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Hiperlipidemias/etiología , Hiperlipidemias/metabolismo , Masculino , Ratones , Ratones Noqueados , Mutación , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo III/genética , Obesidad/etiología , Obesidad/metabolismo
20.
J Neurosci ; 25(7): 1797-805, 2005 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-15716416

RESUMEN

Temporal lobe epilepsy (TLE) is the most common form of epilepsy, affecting approximately 1-2% of the population. Seizure events resulting from TLE are characterized by aberrant hippocampal mossy fiber sprouting and plastic responses that affect brain function. Seizure susceptibility is modulated by the enzyme tissue plasminogen activator (tPA), the normal physiological role of which includes promotion of synaptic reorganization in the mossy fiber pathway by initiating a proteolytic cascade that cleaves extracellular matrix components and influences neurite extension. tPA is concentrated at and selectively secreted from growth cones during excitatory events. However, the mechanisms underlying tPA release during seizure-induced synaptogenesis are not well understood. We examine here potential roles for the signaling enzyme phospholipase D1 (PLD1), which promotes regulated exocytosis in non-CNS cell types, and which we previously demonstrated increases in expression in hippocampal neurons during seizure-induced mossy fiber sprouting. We now show that overexpression of wild-type PLD1 in cultured neurons promotes tPA release and tPA-dependent neurite extension, whereas overexpression of an inactive PLD1 allele or pharmacological inhibition of PLD1 inhibits tPA release. Similarly, viral delivery of wild-type PLD1 into the hippocampus facilitates tPA secretion and mossy fiber sprouting in a seizure-inducing model, whereas the inactive PLD1 allele inhibits tPA release and elicits blunted and abnormal mossy fiber extension similar to that observed for tPA-/- mice. Together, these findings secretion and thus mossy fiber extension in the setting of elevated suggest that PLD1 functions endogenously to regulate tPA-/- neuronal stimulation, such as that seen in TLE.


Asunto(s)
Epilepsia del Lóbulo Temporal/fisiopatología , Neuritas/fisiología , Fosfolipasa D/fisiología , Activador de Tejido Plasminógeno/metabolismo , Animales , Encéfalo/citología , Encéfalo/enzimología , Células Cultivadas/enzimología , Células Cultivadas/ultraestructura , Convulsivantes/toxicidad , Hipocampo/citología , Hipocampo/enzimología , Ácido Kaínico/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Musgosas del Hipocampo/crecimiento & desarrollo , Fibras Musgosas del Hipocampo/ultraestructura , Neuronas/metabolismo , Fosfolipasa D/deficiencia , Fosfolipasa D/genética , Proteínas Recombinantes de Fusión/fisiología , Virus Sindbis/fisiología , Activador de Tejido Plasminógeno/deficiencia , Activador de Tejido Plasminógeno/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA