Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Prostaglandins Other Lipid Mediat ; 152: 106480, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33172790

RESUMEN

Biologically active glutathione (GSH) conjugates of oxygenated fatty acids comprise a group of pro- and anti-inflammatory lipid mediators. While arachidonic acid (AA)-derived conjugates, as the cysteinyl leukotrienes (cys-LTs) and eoxins (EXs) have pro-inflammatory properties, conjugates in tissue regeneration (CTRs) biosynthesized from docosahexaenoic acid (DHA) exhibit pro-resolving activity. Human platelets express abundant amounts of platelet-type 12-lipoxygenase (pt12-LOX) and leukotriene C4 synthase (LTC4S). However, the only two described GSH conjugates formed by platelets are the AA-derived cys-LTs and the recently reported maresin CTRs (MCTRs). While cys-LTs are biosynthesized in a transcellular mechanism via the action of 5-LOX and LTC4S, MCTR1 is formed by 12-LOX and a yet unidentified GSH S-transferase (GST). Here, we present a novel GSH conjugate formed from AA via the 12-LOX pathway in human platelets. The 12-oxo-glutathione adduct, 12-oxo-10-glutathionyl-5,8,14-eicosatrienoic acid (TOG10), was identified by mass spectrometry using positive electrospray ionization. The structural proposal is supported by fragmentation data of the labeled metabolite obtained after incubation of deuterated AA (AA-d8). In platelets as well as in HEK293 cells stably expressing pt12-LOX, TOG10 biosynthesis was inhibited by the 12-LOX inhibitor ML-355 (5 µM), which confirms the involvement of pt12-LOX. Interestingly, TOG10 was formed independently of LTC4S in platelets. This is in accordance with the observation that the conjugate was also generated by AA-stimulated HEK_12-LOX cells in absence of LTC4S. Nevertheless, TOG10 can also be formed by LTC4S as the biosynthesis in HEK_12-LOX_LTC4S cells was reduced by the specific LTC4S inhibitor TK04a. In summary, TOG10 was identified as a new AA-derived GSH conjugate generated in human platelets via the action of pt12-LOX in combination with a GST.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico , Araquidonato 12-Lipooxigenasa , Plaquetas , Glutatión , Células HEK293 , Humanos , Espectrometría de Masas
2.
Prostaglandins Other Lipid Mediat ; 144: 106350, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31323323

RESUMEN

Glutathione (GSH) conjugates of oxygenated polyunsaturated fatty acids comprise a group of pro-inflammatory and pro-resolving lipid mediators formed in immunocompetent cells. While the pro-inflammatory conjugates such as the cysteinyl leukotrienes (cys-LTs), eoxins (EXs) and five-oxo-GSH conjugate (FOG7) derive from arachidonic acid (AA), the group of conjugates in tissue regeneration (CTRs) such as maresin CTRs (MCTRs), protectin CTRs (PCTRs) and resolvin CTRs (RCTRs) are biosynthesized from docosahexaenoic acid (DHA). Here, we present a gradient UPLC-MS/MS method for the analysis of pro-inflammatory and pro-resolving GSH conjugates using positive electrospray ionization (ESI(+)) and collision-induced fragmentation for unambiguous identification and structural information, and a negative ionization (ESI(-)) mode for quantification of the GSH conjugates. The method was employed to detect GSH conjugates in human platelets and macrophages. MCTRs were detected in platelets upon addition of exogenous docosahexaenoic acid (DHA) and the biosynthesis was independent on leukotriene C4 (LTC4) synthase activity. Pathogenic bacteria stimulated the formation of EXs and PCTRs in M2 macrophages, whereas Ca2+-ionophore activated the biosynthesis of LTC4 in M1 and M2 macrophage phenotypes. Together, our methodology covers the qualitative and quantitative analysis of GSH conjugates and gives an analytical basis for the detection and structural elucidation of cysteinyl-containing lipid mediators.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Ácidos Grasos Insaturados/análisis , Glutatión/metabolismo , Oxígeno/metabolismo , Espectrometría de Masas en Tándem/métodos , Plaquetas/metabolismo , Ácidos Grasos Insaturados/sangre , Ácidos Grasos Insaturados/aislamiento & purificación , Ácidos Grasos Insaturados/metabolismo , Humanos , Macrófagos/metabolismo , Fenotipo , Extracción en Fase Sólida
3.
Biochim Biophys Acta ; 1861(11): 1605-1613, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27477678

RESUMEN

Cysteinyl leukotrienes (cys-LTs) cause bronchoconstriction in anaphylaxis and asthma. They are formed by 5-lipoxygenase (5-LOX) from arachidonic acid (AA) yielding the unstable leukotriene A4 (LTA4) that is subsequently conjugated with glutathione (GSH) by LTC4 synthase (LTC4S). Cys-LT receptor antagonists and LTC4S inhibitors have been developed, but only the former have reached the market. High structural homology to related enzymes and lack of convenient test systems due to instability of added LTA4 have hampered the development of LTC4S inhibitors. We present smart cell-free and cell-based assay systems based on in situ-generated LTA4 that allow studying LTC4S activity and investigating LTC4S inhibitors. Co-incubations of microsomes from HEK293 cells expressing LTC4S with isolated 5-LOX efficiently converted exogenous AA to LTC4 (~1.3µg/200µg protein). Stimulation of HEK293 cells co-expressing 5-LOX and LTC4S with Ca2+-ionophore A23187 and 20µM AA resulted in strong LTC4 formation (~250ng/106 cells). MK-886, a well-known 5-LOX activating protein (FLAP) inhibitor that also acts on LTC4S, consistently inhibited LTC4 formation in all assay types (IC50=3.1-3.5µM) and we successfully confirmed TK04a as potent LTC4S inhibitor in these assay systems (IC50=17 and 300nM, respectively). We demonstrated transcellular LTC4 biosynthesis between neutrophils or 5-LOX-expressing HEK293 cells that produce LTA4 from AA and HEK293 cells expressing LTC4S that transform LTA4 to LTC4. In conclusion, our assay approaches are advantageous as the substrate LTA4 is generated in situ and are suitable for studying enzymatic functionality of LTC4S including site-directed mutations and evaluation of LTC4S inhibitors.


Asunto(s)
Bioensayo/métodos , Inhibidores Enzimáticos/farmacología , Glutatión Transferasa/antagonistas & inhibidores , Glutatión Transferasa/metabolismo , Araquidonato 5-Lipooxigenasa/metabolismo , Sistema Libre de Células , Cromatografía Líquida de Alta Presión , Células HEK293 , Humanos , Leucotrieno C4/metabolismo , Microsomas/efectos de los fármacos , Microsomas/metabolismo , Unión Proteica/efectos de los fármacos , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Espectrometría de Masas en Tándem
4.
ChemMedChem ; 17(22): e202200327, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36111583

RESUMEN

We identified 2,4-dinitro-biphenyl-based compounds as new inhibitors of leukotriene C4 synthase (LTC4 S) and 5-lipoxygenase-activating protein (FLAP), both members of the "Membrane Associated Proteins in Eicosanoid and Glutathione metabolism" (MAPEG) family involved in the biosynthesis of pro-inflammatory eicosanoids. By molecular docking we evaluated the putative binding against the targets of interest, and by applying cell-free and cell-based assays we assessed the inhibition of LTC4 S and FLAP by the small molecules at low micromolar concentrations. The present results integrate the previously observed inhibitory profile of the tested compounds against another MAPEG member, i. e., microsomal prostaglandin E2 synthase (mPGES)-1, suggesting that the 2,4-dinitro-biphenyl scaffold is a suitable molecular platform for a multitargeting approach to modulate pro-inflammatory mediators in inflammation and cancer treatment.


Asunto(s)
Compuestos de Bifenilo , Glutatión Transferasa , Simulación del Acoplamiento Molecular , Proteínas Activadoras de la 5-Lipooxigenasa , Compuestos de Bifenilo/farmacología , Prostaglandina-E Sintasas/metabolismo
5.
Cell Chem Biol ; 26(1): 60-70.e4, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30415966

RESUMEN

5-Lipoxygenase (5-LO) initiates the biosynthesis of pro-inflammatory leukotrienes from arachidonic acid, which requires the nuclear membrane-bound 5-LO-activating protein (FLAP) for substrate transfer. Here, we identified human 5-LO as a molecular target of melleolides from honey mushroom (Armillaria mellea). Melleolides inhibit 5-LO via an α,ß-unsaturated aldehyde serving as Michael acceptor for surface cysteines at the substrate entrance that are revealed as molecular determinants for 5-LO activity. Experiments with 5-LO mutants, where select cysteines had been replaced by serine, indicated that the investigated melleolides suppress 5-LO product formation via two distinct modes of action: (1) by direct interference with 5-LO activity involving two or more of the cysteines 159, 300, 416, and 418, and (2) by preventing 5-LO/FLAP assemblies involving selectively Cys159 in 5-LO. Interestingly, replacement of Cys159 by serine prevented 5-LO/FLAP assemblies as well, implying Cys159 as determinant for 5-LO/FLAP complex formation at the nuclear membrane required for leukotriene biosynthesis.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Armillaria/química , Cisteína/metabolismo , Inhibidores de la Lipooxigenasa/farmacología , Sesquiterpenos/farmacología , Células A549 , Relación Dosis-Respuesta a Droga , Humanos , Inhibidores de la Lipooxigenasa/química , Estructura Molecular , Sesquiterpenos/química , Relación Estructura-Actividad
6.
Eur J Med Chem ; 156: 815-830, 2018 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-30053720

RESUMEN

Leukotrienes (LTs) and prostaglandin (PG)E2, produced by 5-lipoxygenase (5-LO) and microsomal prostaglandin E2 synthase-1 (mPGES-1), respectively, are key players in inflammation, and pharmacological suppression of these lipid mediators (LM) represents a strategy to intervene with inflammatory disorders. Previous studies revealed that the benzenesulfonamide scaffold displays efficient 5-LO-inhibitory properties. Here, we structurally optimized benzenesulfonamides which led to an N-phenylbenzenesulfonamide derivative (compound 47) with potent inhibitory activities (IC50 = 2.3 and 0.4 µM for isolated 5-LO and 5-LO in intact cells, respectively). Compound 47 prevented the interaction of 5-LO with its activating protein (FLAP) at the nuclear envelope in transfected HEK293 cells as shown by in situ proximity ligation assay. Comprehensive assessment of the LM profile produced by human macrophages revealed the ability of 47 to selectively down-regulate pro-inflammatory LMs (i.e. LTs and PGE2) in M1 but to enhance the formation of pro-resolving LMs (i.e. resolvins and maresins) in M2 macrophages. Moreover, 47 strongly inhibited LT formation and cell infiltration in two in vivo models of acute inflammation (i.e., peritonitis and air pouch sterile inflammation in mice). Together, 47 represents a novel LT biosynthesis inhibitor with an attractive pharmacological profile as anti-inflammatory drug that also promotes the biosynthesis of pro-resolving LM.


Asunto(s)
Antiinflamatorios/farmacología , Araquidonato 5-Lipooxigenasa/metabolismo , Inhibidores de la Lipooxigenasa/farmacología , Prostaglandina-E Sintasas/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Antiinflamatorios/química , Células Cultivadas , Células HEK293 , Humanos , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/metabolismo , Inhibidores de la Lipooxigenasa/química , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Macrófagos/metabolismo , Masculino , Ratones , Simulación del Acoplamiento Molecular , Prostaglandina-E Sintasas/metabolismo , Sulfonamidas/química , Bencenosulfonamidas
7.
Sci Rep ; 7(1): 9398, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28839250

RESUMEN

Arachidonic acid (AA) is metabolized to diverse bioactive lipid mediators. Whereas the 5-lipoxygenase-activating protein (FLAP) facilitates AA conversion by 5-lipoxygenase (5-LOX) to pro-inflammatory leukotrienes (LTs), the soluble epoxide hydrolase (sEH) degrades anti-inflammatory epoxyeicosatrienoic acids (EETs). Accordingly, dual FLAP/sEH inhibition might be advantageous drugs for intervention of inflammation. We present the in vivo pharmacological profile and efficiency of N-[4-(benzothiazol-2-ylmethoxy)-2-methylphenyl]-N'-(3,4-dichlorophenyl)urea (diflapolin) that dually targets FLAP and sEH. Diflapolin inhibited 5-LOX product formation in intact human monocytes and neutrophils with IC50 = 30 and 170 nM, respectively, and suppressed the activity of isolated sEH (IC50 = 20 nM). Characteristic for FLAP inhibitors, diflapolin (I) failed to inhibit isolated 5-LOX, (II) blocked 5-LOX product formation in HEK cells only when 5-LOX/FLAP was co-expressed, (III) lost potency in intact cells when exogenous AA was supplied, and (IV) prevented 5-LOX/FLAP complex assembly in leukocytes. Diflapolin showed target specificity, as other enzymes related to AA metabolism (i.e., COX1/2, 12/15-LOX, LTA4H, LTC4S, mPGES1, and cPLA2) were not inhibited. In the zymosan-induced mouse peritonitis model, diflapolin impaired vascular permeability, inhibited cysteinyl-LTs and LTB4 formation, and suppressed neutrophil infiltration. Diflapolin is a highly active dual FLAP/sEH inhibitor in vitro and in vivo with target specificity to treat inflammation-related diseases.


Asunto(s)
Inhibidores de Proteína Activante de 5-Lipoxigenasa/farmacología , Inhibidores Enzimáticos/farmacología , Epóxido Hidrolasas/antagonistas & inhibidores , Inhibidores de Proteína Activante de 5-Lipoxigenasa/química , Proteínas Activadoras de la 5-Lipooxigenasa/metabolismo , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Permeabilidad Capilar/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/química , Epóxido Hidrolasas/metabolismo , Humanos , Masculino , Ratones , Estructura Molecular , Transporte de Proteínas
8.
Biochem Pharmacol ; 119: 17-26, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27592027

RESUMEN

The pro-inflammatory leukotrienes (LTs) are formed from arachidonic acid (AA) in activated leukocytes, where 5-lipoxygenase (5-LO) translocates to the nuclear envelope to assemble a functional complex with the integral nuclear membrane protein 5-LO-activating protein (FLAP). FLAP, a MAPEG family member, facilitates AA transfer to 5-LO for efficient conversion, and LT biosynthesis critically depends on FLAP. Here we show that the novel LT biosynthesis inhibitor BRP-187 prevents the 5-LO/FLAP interaction at the nuclear envelope of human leukocytes without blocking 5-LO nuclear redistribution. BRP-187 inhibited 5-LO product formation in human monocytes and polymorphonuclear leukocytes stimulated by lipopolysaccharide plus N-formyl-methionyl-leucyl-phenylalanine (IC50=7-10nM), and upon activation by ionophore A23187 (IC50=10-60nM). Excess of exogenous AA markedly impaired the potency of BRP-187. Direct 5-LO inhibition in cell-free assays was evident only at >35-fold higher concentrations, which was reversible and not improved under reducing conditions. BRP-187 prevented A23187-induced 5-LO/FLAP complex assembly in leukocytes but failed to block 5-LO nuclear translocation, features that were shared with the FLAP inhibitor MK886. Whereas AA release, cyclooxygenases and related LOs were unaffected, BRP-187 also potently inhibited microsomal prostaglandin E2 synthase-1 (IC50=0.2µM), another MAPEG member. In vivo, BRP-187 (10mg/kg) exhibited significant effectiveness in zymosan-induced murine peritonitis, suppressing LT levels in peritoneal exudates as well as vascular permeability and neutrophil infiltration. Together, BRP-187 potently inhibits LT biosynthesis in vitro and in vivo, which seemingly is caused by preventing the 5-LO/FLAP complex assembly and warrants further preclinical evaluation.


Asunto(s)
Proteínas Activadoras de la 5-Lipooxigenasa/metabolismo , Araquidonato 5-Lipooxigenasa/metabolismo , Isoxazoles/farmacología , Antagonistas de Leucotrieno/farmacología , Leucotrienos/biosíntesis , Quinolinas/farmacología , Proteínas Activadoras de la 5-Lipooxigenasa/genética , Animales , Araquidonato 5-Lipooxigenasa/genética , Sistema Libre de Células , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Hidroxiurea/análogos & derivados , Hidroxiurea/farmacología , Indoles/farmacología , Isoxazoles/química , Isoxazoles/metabolismo , Antagonistas de Leucotrieno/química , Antagonistas de Leucotrieno/metabolismo , Masculino , Ratones , Estructura Molecular , Peritonitis/inducido químicamente , Peritonitis/tratamiento farmacológico , Quinolinas/química , Quinolinas/metabolismo , Zimosan/toxicidad
9.
Biochem Pharmacol ; 112: 60-71, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27157409

RESUMEN

5-Lipoxygenase (5-LO) catalyzes the first two steps in leukotriene (LT) biosynthesis. Because LTs play pivotal roles in allergy and inflammation, 5-LO represents a valuable target for anti-inflammatory drugs. Here, we investigated the molecular mechanism, the pharmacological profile, and the in vivo effectiveness of the novel 1,2-benzoquinone-featured 5-LO inhibitor RF-22c. Compound RF-22c potently inhibited 5-LO product synthesis in neutrophils and monocytes (IC50⩾22nM) and in cell-free assays (IC50⩾140nM) without affecting 12/15-LOs, cyclooxygenase (COX)-1/2, or arachidonic acid release, in a specific and reversible manner, supported by molecular docking data. Antioxidant or iron-chelating properties were not evident for RF-22c and 5-LO-regulatory cofactors like Ca(2+) mobilization, ERK-1/2 activation, and 5-LO nuclear membrane translocation and interaction with 5-LO-activating protein (FLAP) were unaffected. RF-22c (0.1mg/kg; i.p.) impaired (I) bronchoconstriction in ovalbumin-sensitized mice challenged with acetylcholine, (II) exudate formation in carrageenan-induced paw edema, and (III) zymosan-induced leukocyte infiltration in air pouches. Taken together, RF-22c is a highly selective and potent 5-LO inhibitor in intact human leukocytes with pronounced effectiveness in different models of inflammation that warrants further preclinical analysis of this agent as anti-inflammatory drug.


Asunto(s)
Antiinflamatorios/farmacología , Araquidonato 5-Lipooxigenasa/metabolismo , Benzoquinonas/farmacología , Broncoconstricción/efectos de los fármacos , Leucotrienos/biosíntesis , Inhibidores de la Lipooxigenasa/farmacología , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/uso terapéutico , Benzoquinonas/administración & dosificación , Benzoquinonas/uso terapéutico , Plaquetas/efectos de los fármacos , Plaquetas/enzimología , Plaquetas/inmunología , Broncoconstricción/inmunología , Células Cultivadas , Edema/tratamiento farmacológico , Edema/enzimología , Edema/inmunología , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Femenino , Humanos , Inhibidores de la Lipooxigenasa/administración & dosificación , Inhibidores de la Lipooxigenasa/uso terapéutico , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Monocitos/efectos de los fármacos , Monocitos/enzimología , Monocitos/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Neutrófilos/inmunología
10.
J Med Chem ; 56(22): 9031-44, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24171493

RESUMEN

Dual inhibition of microsomal prostaglandin E2 synthase-1 (mPGES-1) and 5-lipoxygenase (5-LO) is currently pursued as potential pharmacological strategy for treatment of inflammation and cancer. Here we present a series of 26 novel 2-aminothiazole-featured pirinixic acid derivatives as dual 5-LO/mPGES-1 inhibitors with improved potency (exemplified by compound 16 (2-[(4-chloro-6-{[4-(naphthalen-2-yl)-1,3-thiazol-2-yl]amino}pyrimidin-2-yl)sulfanyl]octanoic acid) with IC50 = 0.3 and 0.4 µM, respectively) and bioactivity in vivo. Computational analysis presumes binding sites of 16 at the tip of the 5-LO catalytic domain and within a subpocket of the mPGES-1 active site. Compound 16 (10 µM) hardly suppressed cyclooxygenase (COX)-1/2 activities, failed to inhibit 12/15-LOs, and is devoid of radical scavenger properties. Finally, compound 16 reduced vascular permeability and inflammatory cell infiltration in a zymosan-induced mouse peritonitis model accompanied by impaired levels of cysteinyl-leukotrienes and prostaglandin E2. Together, 2-aminothiazole-featured pirinixic acids represent potent dual 5-LO/mPGES-1 inhibitors with an attractive pharmacological profile as anti-inflammatory drugs.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Microsomas/enzimología , Pirimidinas/química , Pirimidinas/farmacología , Tiazoles/química , Animales , Araquidonato 5-Lipooxigenasa/química , Sitios de Unión , Diseño de Fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Concentración 50 Inhibidora , Oxidorreductasas Intramoleculares/química , Inhibidores de la Lipooxigenasa/química , Inhibidores de la Lipooxigenasa/metabolismo , Inhibidores de la Lipooxigenasa/farmacocinética , Inhibidores de la Lipooxigenasa/farmacología , Masculino , Ratones , Modelos Moleculares , Peritonitis/inducido químicamente , Peritonitis/tratamiento farmacológico , Prostaglandina-E Sintasas , Conformación Proteica , Pirimidinas/metabolismo , Pirimidinas/farmacocinética , Relación Estructura-Actividad , Zimosan/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA