Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biomolecules ; 14(6)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38927093

RESUMEN

Special attention is given to cow's milk and its variants, with ongoing discussions about health-related impacts primarily focusing on the A1 variant in contrast to the A2 variant. The difference between these variants lies in a single amino acid alteration at position 67 of ß-casein. This alteration is presumed to make the A1 variant more susceptible to enzymatic breakdown during milk digestion, leading to an increased release of the peptide ß-casomorphin-7 (BCM-7). BCM-7 is hypothesized to interact with µ-opioid receptors on immune cells in humans. Although BCM-7 has demonstrated both immunosuppressive and inflammatory effects, its direct impact on the immune system remains unclear. Thus, we examined the influence of A1 and A2 milk on Concanavalin A (ConA)-stimulated human peripheral blood mononuclear cells (PBMCs), as well as the effect of experimentally digested A1 and A2 milk, containing different amounts of free BCM-7 from ß-casein cleavage. Additionally, we evaluated the effects of pure BCM-7 on the proliferation of ConA-stimulated PBMCs and purified CD4+ T cells. Milk fundamentally inhibited PBMC proliferation, independent of the ß-casein variant. In contrast, experimentally digested milk of both variants and pure BCM-7 showed no influence on the proliferation of PBMCs or isolated CD4+ T cells. Our results indicate that milk exerts an anti-inflammatory effect on PBMCs, regardless of the A1 or A2 ß-casein variant, which is nullified after in vitro digestion. Consequently, we deem BCM-7 unsuitable as a biomarker for food-induced inflammation.


Asunto(s)
Caseínas , Proliferación Celular , Endorfinas , Leucocitos Mononucleares , Leche , Fragmentos de Péptidos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/citología , Proliferación Celular/efectos de los fármacos , Leche/química , Endorfinas/farmacología , Endorfinas/metabolismo , Animales , Caseínas/farmacología , Caseínas/metabolismo , Fragmentos de Péptidos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/citología , Concanavalina A/farmacología , Bovinos
2.
J Med Chem ; 66(17): 12396-12406, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37587416

RESUMEN

Noncanonical G protein activation and inactivation, particularly for the Gαi/s protein subfamilies, have long been a focus of chemical research. Combinatorial libraries were already effectively applied to identify modulators of the guanine-nucleotide exchange, as can be exemplified with peptides such as KB-752 and GPM-1c/d, the so-called guanine-nucleotide exchange modulators. In this study, we identified novel bicyclic peptides from a combinatorial library screening that show prominent properties as molecular switch-on/off modulators of Gαi signaling. Among the series of hits, the exceptional paradigm of GPM-3, a protein and state-specific bicyclic peptide, is the first chemically identified GAP (GTPase-activating protein) modulator with a high binding affinity for Gαi protein. Computational analyses identified and assessed the structure of the bicyclic peptides, novel ligand-protein interaction sites, and their subsequent impact on the nucleotide binding site. This approach can therefore lead the way for the development of efficient chemical biological probes targeting Gαi protein modulation within a cellular context.


Asunto(s)
Nucleótidos de Guanina , Biblioteca de Péptidos , Sitios de Unión , Nucleótidos , Guanina
3.
ACS Chem Biol ; 17(2): 463-473, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35042325

RESUMEN

Chemical probes that specifically modulate the activity of heterotrimeric G proteins provide excellent tools for investigating G protein-mediated cell signaling. Herein, we report a family of selective peptidyl Gαi/s modulators derived from peptide library screening and optimization. Conjugation to a cell-penetrating peptide rendered the peptides cell-permeable and biologically active in cell-based assays. The peptides exhibit potent guanine-nucleotide exchange modulator-like activity toward Gαi and Gαs. Molecular docking and dynamic simulations revealed the molecular basis of the protein-ligand interactions and their effects on GDP binding. This study demonstrates the feasibility of developing direct Gαi/s modulators and provides a novel chemical probe for investigating cell signaling through GPCRs/G proteins.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas , Nucleótidos , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Proteínas de Unión al GTP Heterotriméricas/farmacología , Simulación del Acoplamiento Molecular , Nucleótidos/metabolismo , Péptidos/química , Transducción de Señal
4.
Vet Sci ; 10(1)2022 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-36669006

RESUMEN

Enrofloxacin has been shown to be appropriate to treat bacterial eye infections in mammals. However, the anatomy and physiology of the avian eye substantially differ from those in mammals, and pharmacokinetic data substantiating the clinical efficacy of enrofloxacin in birds are still lacking. In total, 40 chickens (Gallus gallus, Lohman Selected Leghorn) received single intramuscular administration of enrofloxacin at a dosage of 25 mg/kg body weight (BW). Serial blood and aqueous humour samples were taken at 12 different time points after administration (0-60 min and 2-32 h) and were analysed for their fluoroquinolone concentrations using a competitive enzyme immunoassay. The metabolization of enrofloxacin to ciprofloxacin was determined using liquid chromatography-mass spectrometry. The maximum serum concentrations of fluoroquinolones were observed at the time point of 2.82 ± 0.1 h and amounted to 10.67 ± 0.5 µg/mL. Fluoroquinolones redistributed to a minor extent into the aqueous humour reaching maximum concentrations of 4.52 ± 1.2 µg/mL after 7.54 ± 1.0 h of drug administration. The mean residence time (MRT), volume of distribution (Vd), and terminal half-life (t1/2 ß) were 1.68-, 2.84-, and 2.01-fold higher in aqueous humour than in serum, indicating that fluoroquinolones were trapped in aqueous humour. Enrofloxacin was only marginally metabolized into ciprofloxacin. A single intramuscular injection of a therapeutical dose of enrofloxacin (25 mg/kg BW) thus generated sustained and therapeutically active levels of enrofloxacin in the aqueous humour of chicken eyes.

8.
Cell Signal ; 25(12): 2440-52, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23993961

RESUMEN

The tumor suppressor Fhit protein is defective or absent in many tumor cells due to methylation, mutation or deletion of the FHIT gene. Despite numerous attempts to unravel the functions of Fhit, the mechanisms by which the function and expression of Fhit are regulated remain poorly understood. We have recently shown that activated Gαq subunits interact directly with Fhit and enhance its inhibitory effect on cell growth. Here we investigated the regulation of Fhit expression by Gq. Our results showed that Fhit was up-regulated specifically by activating Gα subunits of the Gq subfamily but not by those of the other G protein subfamilies. This up-regulation effect was mediated by a PKC/MEK pathway independent of Src-mediated Fhit Tyr(114) phosphorylation. We further demonstrated that elevated Fhit expression was due to the specific regulation of Fhit protein synthesis in the ribosome by activated Gαq, where the regulations of cap-dependent protein synthesis were apparently not required. Moreover, we showed that activated Gαq could increase cell-cell adhesion through Fhit. These findings provide a possible handle to modulate the level of the Fhit tumor suppressor by manipulating the activity of Gq-coupled receptors.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Proteínas de Neoplasias/genética , Regulación hacia Arriba , Ácido Anhídrido Hidrolasas/metabolismo , Línea Celular , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas de Neoplasias/metabolismo , Biosíntesis de Proteínas , Proteína Quinasa C/metabolismo , Transducción de Señal
9.
Cell Commun Signal ; 11: 59, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23947369

RESUMEN

BACKGROUND: The FHIT tumor suppressor gene is arguably the most commonly altered gene in cancer since it is inactivated in about 60% of human tumors. The Fhit protein is a member of the ubiquitous histidine triad proteins which hydrolyze dinucleoside polyphosphates such as Ap3A. Despite the fact that Fhit functions as a tumor suppressor, the pathway through which Fhit inhibits growth of cancer cells remains largely unknown. Phosphorylation by Src tyrosine kinases provides a linkage between Fhit and growth factor signaling. Since many G proteins can regulate cell proliferation through multiple signaling components including Src, we explored the relationship between Gα subunits and Fhit. RESULTS: Several members of the Gαq subfamily (Gα16, Gα14, and Gαq) were found to co-immunoprecipitate with Fhit in their GTP-bound active state in HEK293 cells. The binding of activated Gαq members to Fhit appeared to be direct and was detectable in native DLD-1 colon carcinoma cells. The use of Gα16/z chimeras further enabled the mapping of the Fhit-interacting domain to the α2-ß4 region of Gα16. However, Gαq/Fhit did not affect either Ap3A binding and hydrolysis by Fhit, or the ability of Gαq/16 to regulate downstream effectors including phospholipase Cß, Ras, ERK, STAT3, and IKK. Functional mutants of Fhit including the H96D, Y114F, L25W and L25W/I10W showed comparable abilities to associate with Gαq. Despite the lack of functional regulation of Gq signaling by Fhit, stimulation of Gq-coupled receptors in HEK293 and H1299 cells stably overexpressing Fhit led to reduced cell proliferation, as opposed to an enhanced cell proliferation typically seen with parental cells. CONCLUSIONS: Activated Gαq members interact with Fhit through their α2-ß4 region which may result in enhancement of the growth inhibitory effect of Fhit, thus providing a possible avenue for G protein-coupled receptors to modulate tumor suppression.


Asunto(s)
Ácido Anhídrido Hidrolasas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ácido Anhídrido Hidrolasas/genética , Calcio/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Fosfatos de Dinucleósidos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Humanos , Fosfatos de Inositol/metabolismo , Mutación , Proteínas de Neoplasias/genética , Fosforilación , ARN Interferente Pequeño/genética , Proteínas Supresoras de Tumor/genética , Familia-src Quinasas/metabolismo
10.
PLoS One ; 8(6): e68168, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840827

RESUMEN

Protein S-nitrosylation, the covalent attachment of a nitroso moiety to thiol groups of specific cysteine residues, is one of the major pathways of nitric oxide signaling. Hundreds of proteins are subject to this transient post-translational modification and for some the functional consequences have been identified. Biochemical assays for the analysis of protein S-nitrosylation have been established and can be used to study if and under what conditions a given protein is S-nitrosylated. In contrast, the equally desirable subcellular localization of specific S-nitrosylated protein isoforms has not been achieved to date. In the current study we attempted to specifically localize S-nitrosylated α- and ß-tubulin isoforms in primary neurons after fixation. The approach was based on in situ replacement of the labile cysteine nitroso modification with a stable tag and the subsequent use of antibodies which recognize the tag in the context of the tubulin polypeptide sequence flanking the cysteine residue of interest. We established a procedure for tagging S-nitrosylated proteins in cultured primary neurons and obtained polyclonal anti-tag antibodies capable of specifically detecting tagged proteins on immunoblots and in fixed cells. However, the antibodies were not specific for tubulin isoforms. We suggest that different tagging strategies or alternative methods such as fluorescence resonance energy transfer techniques might be more successful.


Asunto(s)
Anticuerpos/metabolismo , Inmunohistoquímica/métodos , S-Nitrosotioles/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Células Cultivadas , Cisteína/metabolismo , Ratones , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Isoformas de Proteínas/metabolismo , Compuestos de Sulfhidrilo/metabolismo
11.
PLoS One ; 8(1): e53510, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23308242

RESUMEN

BACKGROUND: There is increasing evidence that opioid analgesics may interfere with tumour growth. It is currently thought that these effects are mediated by transactivation of receptor tyrosine kinase (RTK)-controlled ERK1/2 and Akt signalling. The growth of many breast cancer cells is dependent on hyperactive ErbB receptor networks and one of the most successful approaches in antineoplastic therapy during the last decade was the development of ErbB-targeted therapies. However, the response rates of single therapies are often poor and resistance mechanisms evolve rapidly. To date there is no information about the ability of opioid analgesics to interfere with the growth of ErbB-driven cancers. METHODS AND PRINCIPAL FINDINGS: Here we demonstrate that ErbB2 overexpressing BT474 human breast cancer cells carry fully functional endogenous µ-opioid receptors. Most interestingly, the acute opioid effects on basal and Heregulin-stimulated ERK1/2 and Akt phosphorylation changed considerably during chronic Morphine treatment. Investigation of the underlying mechanism by the use of protein kinase inhibitors and co-immunoprecipitation studies revealed that chronic Morphine treatment results in rearrangement of the ErbB signalling network leading to dissociation of ERK1/2 from Akt signalling and a switch from ErbB1/ErbB3 to ErbB1/ErbB2-dependent cell growth. In chronically Morphine-treated cells Heregulin-stimulated ERK1/2 signalling is redirected via a newly established PI3K- and metalloproteinase-dependent feedback loop. Together, these alterations result in apoptosis of BT474 cells. A similar switch in Heregulin-stimulated ERK1/2 signalling from an ErbB2-independent to an ErbB2-, PI3K- and metalloproteinase-dependent mechanism was also observed in κ-opioid receptor expressing SKBR3 human mammary adenocarcinoma cells. CONCLUSIONS AND SIGNIFICANCE: The present data demonstrate that the ErbB receptor network of human breast cancer cells represents a target for chronic Morphine treatment. Rearrangement of ErbB signalling by chronic Morphine may provide a promising strategy to enhance the sensitivity of breast cancer cells to ErbB-directed therapies and to prevent the development of escape mechanisms.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Morfina/farmacología , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Transducción de Señal/efectos de los fármacos , Adenocarcinoma/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Humanos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neurregulina-1/genética , Neurregulina-1/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
12.
Berl Munch Tierarztl Wochenschr ; 125(11-12): 520-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23227771

RESUMEN

A central nervous disorder occurred spontaneously in a herd of feeder pigs characterized by muscle fasciculations, convulsions, squealing, and acute death in numerous animals. Histopathology revealed a degenerative poliomyeloencephalopathy of brain stem and spinal cord consisting of neuronal hypertrophy, chromatolysis, neuronophagia, and satellitosis associated with Wallerian degeneration of ventral rootlets and neurogenic muscle atrophy of limb musculature. The sudden onset of clinical signs and the pattern of morphological findings were suggestive of intoxication. Though parathion was found in two animals, serum acetylcholine esterase activity and morphological findings were not compatible with an organophosphate poisoning. A hereditary disorder was excluded by genetic analysis. Summarized findings in the present cases are reminiscent of changes observed in ruminants suffering from patulin poisoning, a neuromycotoxicosis caused by Aspergillus clavatus. However, toxicological and microbiological investigations failed to identify the cause of this unusual and so far not described disease in pigs. Morphologically, lesion distribution and alterations of motor neurons resemble changes observed in equine motor neuron disease, spinal muscular atrophy of certain canine breeds, and amyotrophic lateral sclerosis (Lou Gehrig's disease) in man. Therefore, the term spontaneous porcine motor neuron disease (SPMND) is proposed for this new and unique entitiy.


Asunto(s)
Enfermedad de la Neurona Motora/veterinaria , Enfermedades de los Porcinos/diagnóstico , Animales , Enfermedad de la Neurona Motora/sangre , Porcinos , Enfermedades de los Porcinos/sangre
13.
J Mol Cell Cardiol ; 52(1): 196-205, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22085704

RESUMEN

Endothelial hyperpermeability followed by edema formation is a hallmark of many severe disorders. Effective drugs directly targeting endothelial barrier function are widely lacking. We hypothesized that the hawthorn (Crataegus spp.) extract WS® 1442, a proven multi-component drug against moderate forms of heart failure, would prevent vascular leakage by affecting endothelial barrier-regulating systems. In vivo, WS® 1442 inhibited the histamine-evoked extravasation of FITC-dextran from mouse cremaster muscle venules. In cultured human endothelial cells, WS® 1442 blocked the thrombin-induced FITC-dextran permeability. By applying biochemical and microscopic techniques, we revealed that WS® 1442 abrogates detrimental effects of thrombin on adherens junctions (vascular endothelial-cadherin), the F-actin cytoskeleton, and the contractile apparatus (myosin light chain). Mechanistically, WS® 1442 inhibited the thrombin-induced rise of intracellular calcium (ratiometric measurement), followed by an inactivation of PKC and RhoA (pulldown assay). Moreover, WS® 1442 increased endothelial cAMP levels (ELISA), which consequently activated PKA and Rap1 (pulldown assay). Utilizing pharmacological inhibitors or siRNA, we found that PKA is not involved in barrier protection, whereas Epac1, Rap1, and Rac1 play a crucial role in the WS® 1442-induced activation of cortactin, which triggers a strong cortical actin rearrangement. In summary, WS® 1442 effectively protects against endothelial barrier dysfunction in vitro and in vivo. It specifically interacts with endothelial permeability-regulating systems by blocking the Ca(2+)/PKC/RhoA and activating the cAMP/Epac1/Rap1 pathway. As a proven safe herbal drug, WS® 1442 opens a novel pharmacological approach to treat hyperpermeability-associated diseases. This in-depth mechanistic work contributes to a better acceptance of this herbal remedy.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , AMP Cíclico/metabolismo , Células Endoteliales/metabolismo , Flavonoides/farmacología , Extractos Vegetales/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas de Unión al GTP rap1/metabolismo , Uniones Adherentes/efectos de los fármacos , Animales , Calcio/metabolismo , Células Cultivadas , Cortactina/metabolismo , Crataegus , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa C/metabolismo , Fibras de Estrés/efectos de los fármacos , Trombina/farmacología , Proteína de Unión al GTP rhoA/metabolismo
14.
Mol Pharmacol ; 79(2): 326-35, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21078885

RESUMEN

δ-Opioid receptor (DOR)-induced activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) is mediated by the transactivation of epidermal growth factor (EGF) receptors. Here we demonstrate that in stably DOR-expressing human embryonic kidney (HEK) 293 (HEK/DOR) cells, down-regulation of EGF receptors by long-term EGF (0.1 µg for 18 h) treatment, but not by small interfering RNA, results in functional desensitization of EGF (10 ng/ml)-stimulated ERK1/2 signaling. In EGF receptor-desensitized (HEK/DOR(-EGFR)) cells, however, [d-Ala²,d-Leu5]enkephalin (1 µM) and etorphine (0.1 µM) retained their ability to stimulate ERK1/2 activation. The newly acquired signal transduction mechanism is insensitive to the EGF receptor blockers 4-(3-chloroanilino)-6,7-dimethoxyquinazoline (AG1478) and N-[4-[(3-bromophenyl)amino]-6-quinazolinyl]-2-butynamide (CL-387,785), does not involve DOR internalization and activation of the focal adhesion kinase pp125FAK, but requires matrix metalloproteinase-dependent release of soluble growth factors. A supernatant transfer assay in which conditioned growth media of opioid-treated HEK/DOR and HEK/DOR(-EGFR) "donor" cells are used to stimulate ERK1/2 activity in DOR-lacking HEK293 wild type and HEK293(-EGFR) "acceptor" cells revealed that long-term EGF treatment produces a switch in the receptor tyrosine kinase (RTK) system transactivated by opioids. Using microfluidic electrophoresis, chemical inhibitors, phosphorylation-specific antibodies, and EGF receptor-deficient Chinese hamster ovary-K1 cells, we identified the release of an insulin-like growth factor-1 (IGF-1)-like peptide and activation of IGF-1 receptors in HEK/DOR(-EGFR) cells after DOR activation. A similar switch from a neurotrophic tyrosine kinase receptor type 1 to an IGF-1 receptor-dependent ERK1/2 signaling was observed for chronically nerve growth factor-treated neuroblastoma × glioma (NG108-15) cells. These results indicate that transactivation of the dominant RTK system in a given cellular setting may represent a general feature of opioids to maintain mitogenic signaling.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptores Opioides delta/metabolismo , Western Blotting , Línea Celular , Humanos , Microfluídica , Microscopía Confocal , Fosforilación , Quinazolinas/farmacología , Receptor IGF Tipo 1/genética , Activación Transcripcional , Tirfostinos/farmacología
15.
J Biol Chem ; 284(50): 34819-28, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19828455

RESUMEN

Opioids display ligand-specific differences in the time course of ERK1/2 signaling. Whereas full agonists, like etorphine, induce only transient activation of ERK1/2, the partial agonist morphine mediates persistent stimulation of mitogenic signaling. Here we report that in stably delta-opioid receptor (DOR)-expressing HEK293 (HEK/DOR) cells, the transient nature of etorphine-induced ERK1/2 signaling is due to desensitization of epidermal growth factor (EGF) receptor-mediated activation of the Ras/Raf-1/ERK1/2 cascade. Desensitization of ERK1/2 activity by etorphine is associated with down-regulation of EGF receptors, an effect mediated by the ubiquitin ligase c-Cbl. In contrast, chronic morphine treatment failed to desensitize EGF receptors, resulting in unimpeded ERK1/2 signaling. The failure of morphine to desensitize ERK1/2 signaling is mediated by persistent activation of c-Src, which induces degradation of c-Cbl. The role of c-Src in opioid-specific ERK1/2 signaling is further demonstrated by pretreatment of the cells with PP2 and SKI-I as well as overexpression of a dominant negative c-Src mutant (c-Src(dn)) or a c-Src-resistant c-Cbl mutant (CblY3F), both of which facilitate desensitization of ERK1/2 signaling by morphine. Conversely, overexpression of c-Src as well as down-regulation of c-Cbl by small interfering RNA results in persistent etorphine-induced stimulation of ERK1/2 activity. Subcellular fractionation experiments finally attributed the ability of morphine to persistently activate c-Src to its redistribution from Triton X-100-insensitive membrane rafts to DOR and EGF receptor containing high density membrane compartments implicated in ERK1/2 signaling. These results demonstrate that agonist-specific differences in the temporal and spatial pattern of c-Src activation determine the kinetics of DOR-mediated regulation of ERK1/2 signaling.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfina/farmacología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Receptores Opioides delta/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Línea Celular , Activación Enzimática , Receptores ErbB/metabolismo , Etorfina/farmacología , Humanos , Microdominios de Membrana/metabolismo , Ratones , Narcóticos/farmacología , Proteínas Tirosina Quinasas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Familia-src Quinasas
16.
Exp Cell Res ; 315(12): 2115-25, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19362548

RESUMEN

delta-Opioid receptor (DOR) agonists possess cytoprotective properties, an effect associated with activation of the "pro-survival" kinase Akt. Here we delineate the signal transduction pathway by which opioids induce Akt activation in neuroblastomaxglioma (NG108-15) hybrid cells. Exposure of the cells to both [D-Pen(2,5)]enkephalin and etorphine resulted in a time- and dose-dependent increase in Akt activity, as measured by means of an activation-specific antibody recognizing phosphoserine-473. DOR-mediated Akt signaling is blocked by the opioid antagonist naloxone and involves inhibitory G(i/o) proteins, because pre-treatment with pertussis toxin, but not over-expression of the G(q/11) scavengers EBP50 and GRK2-K220R, prevented this effect. Further studies with Wortmannin and LY294002 revealed that phophoinositol-3-kinase (PI3K) plays a central role in opioid-induced Akt activation. Opioids stimulate Akt activity through transactivation of receptor tyrosine kinases (RTK), because pre-treatment of the cells with inhibitors for neurotrophin receptor tyrosine kinases (AG879) and the insulin-like growth factor receptor IGF-1 (AG1024), but not over-expression of the Gbetagamma scavenger phosducin, abolished this effect. Activated Akt translocates to the nuclear membrane, where it promotes GSK3 phosphorylation and prevents caspase-3 cleavage, two key events mediating inhibition of cell apoptosis and enhancement of cell survival. Taken together, these results demonstrate that in NG108-15 hybrid cells DOR agonists possess cytoprotective properties mediated by activation of the RTK/PI3K/Akt signaling pathway.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Opioides delta/agonistas , Línea Celular Tumoral , Supervivencia Celular , Citoprotección , Encefalinas/farmacología , Activación Enzimática , Etorfina/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Subunidades beta de la Proteína de Unión al GTP/fisiología , Subunidades gamma de la Proteína de Unión al GTP/fisiología , Glioma , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Células Híbridas/metabolismo , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neuroblastoma , Membrana Nuclear/metabolismo , Fosforilación , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/agonistas , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides delta/fisiología , Transducción de Señal , Activación Transcripcional
17.
J Cell Mol Med ; 13(8B): 2122-2130, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19175691

RESUMEN

The standardised Ginkgo biloba extract EGb 761 (Dr. Willmar Schwabe Pharmaceuticals, Karlsruhe, Germany) is one of the most widely used herbal remedies. Indications for this extract range from dementia to peripheral vascular disease, based on well-documented vascular effects. Surprisingly, the actions of EGb 761 on angiogenesis as a function of vascular cells have not been investigated to date. The anti-cancer activity of EGb 761 in vitro and epidemiological data showing reduced risk for ovarian cancer in regular users have prompted us to investigate this issue. We show an anti-angiogenic profile of EGb 761 in vitro (inhibited proliferation, migration and tube formation of endothelial cells) and in vivo in the chicken chorio-allantoic membrane (CAM) assay. An analysis of the underlying mechanisms indicates inhibition of growth factor-induced extracellular signal-regulated kinase (ERK) phosphorylation by EGb 761. Inhibitory effects of EGb 761 on ERK as well as of the upstream kinases map-erk-kinase (MEK) and rapidly growing fibrosarcoma (Raf)-1 could be completely reversed by pre-treatment with sodium vanadate (inhibitor of tyrosine phosphatases). Sodium vanadate also reversed the EGb 761-induced inhibition of endothelial cell migration. Focusing on tyrosine phosphatases upstream of the Raf-MEK-ERK cascade, we identified the tyrosine phosphatase Src homology-2 domain-containing phosphatase 1 (SHP-1) as one target of EGb 761. SHP-1 was rapidly activated by EGb 761, and silencing SHP-1 (siRNA) abrogated reduction of endothelial proliferation by EGb 761. In summary, we identify EGb 761 as a potent anti-angiogenic drug. The underlying mechanism is the activation of protein tyrosine phosphatases, leading to inhibition of the Raf-MEK-ERK pathway. These findings provide a rational basis for using EGb 761 for an additional therapeutic indication: anti-angiogenesis-based tumour prevention and adjuvant therapy.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Extractos Vegetales/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Animales , Secuencia de Bases , Línea Celular Transformada , Activación Enzimática , Femenino , Ginkgo biloba , Humanos , Fosforilación , ARN Interferente Pequeño
18.
Cell Signal ; 20(12): 2324-31, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18804531

RESUMEN

Integrin-mediated cell adherence to extracellular matrix proteins results in stimulation of ERK1/2 activity, a mechanism involving focal adhesion tyrosine kinases (pp125FAK, Pyk-2) and epidermal growth factor receptors (EGFRs). G protein-coupled receptors (GPCRs) may also mediate ERK1/2 activation in an integrin-dependent manner, the underlying signaling mechanism of which still remains unclear. Here we demonstrate that the delta-opioid receptor (DOR), a typical GPCR, stimulates ERK1/2 activity in HEK293 cells via integrin-mediated transactivation of EGFR function. Inhibition of integrin signaling by RGDT peptides, cytochalasin, and by keeping the cells in suspension culture both blocked [D-Ala(2), D-Leu(5)]enkephalin (DADLE)- and etorphine-stimulated ERK1/2 activity. Integrin-dependent ERK1/2 activation does not involve FAK/Pyk-2, because over-expression of the FAK/Pyk-2 inhibitor SOCS-3 failed to attenuate DOR signaling. Exposure of the cells to the EGFR inhibitors AG1478 and BPIQ-I blocked DOR-mediated ERK1/2 activation. Because RGDT peptides also prevented DOR-mediated EGFR activation, the present findings indicate that in HEK293 cells DOR-stimulated ERK1/2 activity is mediated by integrin-stimulated EGFRs. Further studies with the phospholipase C (PLC) inhibitors U73122 and ET-18-OCH(3) revealed that opioid-stimulated integrin activation is sensitive to PLC. In contrast, integrin-mediated transactivation of EGFR function appears to be dependent on PKC-delta, as indicated by studies with rottlerin and siRNA knock-down. A similar ERK1/2 signaling pathway was observed for NG108-15 cells, a neuronal cell line endogenously expressing the DOR. In these cells, the nerve growth factor TrkA receptor replaces the EGFR in connecting DOR-activated integrins to the Ras/Raf/ERK1/2 pathway. Together, these data describe an alternative ERK1/2 signaling pathway in which the DOR transactivates the growth factor receptor associated mitogen-activated protein kinase cascade in an integrin-dependent manner.


Asunto(s)
Receptores ErbB/metabolismo , Integrinas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores Opioides delta/metabolismo , Células Cultivadas , Leucina Encefalina-2-Alanina/farmacología , Receptores ErbB/antagonistas & inhibidores , Estrenos/farmacología , Etorfina/farmacología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Oligopéptidos/farmacología , Fosforilación , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Pirrolidinonas/farmacología , Quinazolinas , Fosfolipasas de Tipo C/metabolismo , Tirfostinos/farmacología
19.
FEBS Lett ; 582(23-24): 3325-9, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18775707

RESUMEN

This study demonstrates that activation of delta-opioid receptors (DORs) in neuroblastomaxglioma (NG108-15) hybrid cells by [D-Ala2, D-Leu5]enkephalin (DADLE) and etorphine significantly enhances cell adhesion to fibronectin-coated wells. This effect is blocked by both naloxone and integrin binding RGDT peptides. In addition, cell adhesion turned out to be a prerequisite for DOR-stimulated transactivation of Tropomyosin-related kinase A (TrkA) and extracellular signal-regulated kinases 1/2 (ERK1/2). Because inhibition of TrkA activation by AG879 completely blocked DOR- and integrin-mediated ERK1/2 signaling, the present results indicate that in NG108-15 cells DOR-stimulated ERK1/2 activation is mediated by integrin-induced transactivation of TrkA.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/enzimología , Receptor trkA/agonistas , Receptores Opioides delta/agonistas , Adhesión Celular , Leucina Encefalina-2-Alanina/farmacología , Etorfina/farmacología , Glioma , Humanos , Células Híbridas/efectos de los fármacos , Células Híbridas/enzimología , Integrinas/metabolismo , Naloxona/farmacología , Neuroblastoma , Neuronas/efectos de los fármacos , Neuronas/fisiología , Oligopéptidos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Receptor trkA/metabolismo , Activación Transcripcional , Tirfostinos/farmacología
20.
Mol Pharmacol ; 66(6): 1719-26, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15448191

RESUMEN

Inflammatory pain is counteracted by a number of physiological processes. For example, opioid receptors, which are present on peripheral terminals of sensory neurons, are activated by endogenous opioids, which are released from immune cells migrating to the inflamed tissue. Earlier data demonstrated that interleukin-6 contributes to such inflammation-induced analgesia. In this report, we demonstrated that interleukin-6 strongly induces mu-opioid receptor mRNA in the human neuroblastoma cell line SH SY5Y, whereas delta-opioid receptor mRNA levels are not influenced. The mRNA increase in these cells is followed by an increase in mu-opioid receptor-specific binding. Using transcription factor decoy oligonucleotides, direct evidence was provided that the up-regulation of mu-opioid receptor mRNA in intact cells is dependent on the transcription factors signal transducers and activators of transcription 1 (STAT1) and STAT3, whereas other transcription factors, such as activator protein-1, nuclear factor (NF)-kappaB, or NF-interleukin-6 are not involved. STAT1 and STAT3 bound to a site located at nucleotide -1583 on the promoter of the human mu-opioid receptor gene, as shown by transient transfection experiments, electrophoretic mobility shift assays, and transcription factor decoy oligonucleotides. A mutation analysis of the 5'-TTCATGGAA-3' STAT1/3 element (palindrome underlined) was performed to determine nucleotide residues that are necessary for the binding of STAT1 and STAT3. It suggested that only the palindromic half sides and the two adjacent central nucleotides are required. Neither mutation of the nucleotides outside the palindrome nor mutation of the central nucleotide affected STAT1/3 binding.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Interleucina-6/farmacología , Receptores Opioides mu/genética , Transcripción Genética/efectos de los fármacos , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación , Neuroblastoma , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides delta/genética , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...