Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Lett Appl Microbiol ; 60(4): 352-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25588530

RESUMEN

UNLABELLED: Newly synthesized erythromycin derivatives were screened for synergy with oxacillin and other ß-lactam antibiotics against methicillin-resistant Staphylococcus aureus (MRSA). MRSA ATCC43300 and some clinically isolated MRSA were used. Several erythromycin derivatives were found to possess high synergism with oxacillin against MRSA. The newly synthesized erythromycin derivatives were also tested for their inhibitory effects against MRSA, either separately or in combination with oxacillin, using serial broth dilution, disc diffusion, Etest strips, growth curves and time-kill curves. A representative derivative, SIPI-8294, could potentiate almost all ß-lactam antibiotics tested against the model strain MRSA ATCC43300 from 4 to 128 times and had synergism with oxacillin against 12 of 16 clinical isolates of MRSA under one-fourth of the minimum inhibitory concentration (MIC) of the compounds. This is the first report on the synergistic activity of these new erythromycin derivatives. These findings provide a new choice for the treatment of infection caused by MRSA and lead us to further study the synergistic mechanism. SIGNIFICANCE AND IMPACT OF THE STUDY: This study is the first report on the synergy of anti-MRSA between new erythromycin derivatives and ß-lactam antibiotics in vitro. The results show that although the erythromycin derivatives have poor anti-MRSA effects alone, they possess high synergism with oxacillin against MRSA ATCC43300 and clinically isolated MRSA. These novel compounds can significantly reduce the dosage of ß-lactam antibiotics against MRSA, while this synergistic effect is different from the combination of ß-lactams and ß-lactamase inhibitors. The research may provide a new choice for the treatment of infection caused by MRSA and be useful to the research and development of new combination of medicines.


Asunto(s)
Antibacterianos/farmacología , Eritromicina/análogos & derivados , Eritromicina/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Oxacilina/farmacología , Farmacorresistencia Bacteriana Múltiple , Sinergismo Farmacológico , Humanos , Pruebas de Sensibilidad Microbiana
2.
Oncogene ; 25(15): 2234-44, 2006 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-16301993

RESUMEN

The membrane redistribution and phosphorylation of focal adhesion kinase (FAK) have been reported to be important for cell migration. We previously showed that Lysophosphatidic acid (LPA) induced FAK membrane redistribution and autophosphorylation in ovarian cancer SK-OV3 cells and the signaling pathway consisting of Gi-Ras-MEKK1 mediated LPA-induced FAK membrane redistribution but not FAK autophosphorylation. We also showed that the disruption of the Gi-Ras-MEKK1 pathway led to a significant reduction in LPA-stimulated cell migration. These findings raised the question of whether LPA-induced FAK autophosphorylation was required for LPA-stimulated cell migration and what signaling mechanism was involved in LPA-induced FAK autophosphorylation. In this study, we expressed the membrane anchored wild-type FAK (CD2-FAK) in SK-OV3 cells and found that the expression of CD2-FAK greatly rescued LPA-stimulated cell migration in Gi or Ras-inhibited cells. However, Gi inhibitor pertussis toxin or dominant-negative H-Ras still significantly inhibited LPA-stimulated cell migration in cells expressing the membrane anchored FAK containing a mutation in the autophosphorylation site [CD2-FAK(Y397A)]. These results suggest that FAK autophosphorylation plays a role in LPA-stimulated cell migration. With the aid of p115RhoGEF-RGS, G12 and G13 minigenes to inhibit G12/13, we found that the G12/13 pathway was required for LPA-induced FAK autophosphorylation and efficient cell migration. Moreover, LPA activated RhoA and Rho kinase (ROCK) in a G12/13-dependent manner and their activities were required for LPA-induced FAK autophosphorylation. However, Rho or ROCK inhibitors displayed no effect on LPA-induced FAK membrane redistribution although they abolished LPA-induced cytoskeleton reorganization. Our studies show that the G12/13-RhoA-ROCK signaling pathway mediates LPA-induced FAK autophosphorylation and contributes to LPA-stimulated cell migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Lisofosfolípidos/farmacología , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Transducción de Señal , Proteína de Unión al GTP rhoA/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Adhesiones Focales/enzimología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Genes ras/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Mutación , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Células Tumorales Cultivadas , Proteínas ras/fisiología , Quinasas Asociadas a rho
3.
J Leukoc Biol ; 70(6): 839-48, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11739545

RESUMEN

Accumulating evidence indicates that G-protein-coupled receptors (GPCRs) play an active role in transcriptional regulation. In leukocytes, activation of receptors for several chemokines and classic chemoattractants has been associated with enhanced expression of proinflammatory cytokines and chemokines. GPCRs in endothelial and epithelial cells also regulate transcription and contribute to the expression of cytokines, adhesion molecules, and growth factors that are essential for extravasation of leukocytes and tissue repair. Nuclear factor (NF) kappaB is one of the most important transcription factors responsible for the expression of these proinflammatory genes. Recent studies have shown that GPCRs utilize several different pathways to activate NF-kappaB. These pathways differ from the ones induced by classic cytokines in that they are initiated by heterotrimeric G-proteins, but they converge to IkappaB phosphorylation and nuclear translocation/modification of the NF-kappaB proteins. GPCR-induced NF-kappaB activation provides an effective means for local expression of cytokine and growth factor genes due to the wide distribution of these receptors. Chemokine-induced, GPCR-mediated production of chemokines constitutes an autocrine regulatory mechanism for the growth of certain malignant tumors and enhances the recruitment of leukocytes to sites of inflammation.


Asunto(s)
Proteínas de Unión al GTP/fisiología , Leucocitos/fisiología , FN-kappa B/fisiología , Receptores de Superficie Celular/fisiología , Transducción de Señal , Animales , Comunicación Autocrina , Humanos , Activación Transcripcional
4.
J Biol Chem ; 276(49): 45979-87, 2001 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-11590141

RESUMEN

The Kaposi's sarcoma herpesvirus (KSHV) open reading frame 74 encodes a G protein-coupled receptor (GPCR) for chemokines. Exogenous expression of this constitutively active GPCR leads to cell transformation and vascular overgrowth characteristic of Kaposi's sarcoma. We show here that expression of KSHV-GPCR in transfected cells results in constitutive transactivation of nuclear factor kappa B (NF-kappa B) and secretion of interleukin-8, and this response involves activation of G alpha(13) and RhoA. The induced expression of a NF-kappa B luciferase reporter was partially reduced by pertussis toxin and the G beta gamma scavenger transducin, and enhanced by co-expression of G alpha(13) and to a lesser extent, G alpha(q). These results indicate coupling of KSHV-GPCR to multiple G proteins for NF-kappa B activation. Expression of KSHV-GPCR led to stress fiber formation in NIH 3T3 cells. To examine the involvement of the G alpha(13)-RhoA pathway in KSHV-GPCR-mediated NF-kappa B activation, HeLa cells were transfected with KSHV-GPCR alone and in combination with the regulator of G protein signaling (RGS) from p115RhoGEF or a dominant negative RhoA(T19N). Both constructs, as well as the C3 exoenzyme from Clostritium botulinum, partially reduced NF-kappa B activation by KSHV-GPCR, and by a constitutively active G alpha(13)(Q226L). KSHV-GPCR-induced NF-kappa B activation is accompanied by increased secretion of IL-8, a function mimicked by the activated G alpha(13) but not by an activated G alpha(q)(Q209L). These results suggest coupling of KSHV-GPCR to the G alpha(13)-RhoA pathway in addition to other G proteins.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al GTP/metabolismo , Herpesvirus Humano 6/metabolismo , Interleucina-8/metabolismo , FN-kappa B/metabolismo , Receptores de Superficie Celular/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Células HeLa , Humanos , Sarcoma de Kaposi/virología , Transducción de Señal
5.
Am J Physiol Lung Cell Mol Physiol ; 281(5): L1037-50, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11597894

RESUMEN

Acute lung injury occurs as a result of a cascade of cellular events initiated by either infectious or noninfectious inflammatory stimuli. An elevated level of proinflammatory mediators combined with a decreased expression of anti-inflammatory molecules is a critical component of lung inflammation. Expression of proinflammatory genes is regulated by transcriptional mechanisms. Nuclear factor-kappa B (NF-kappa B) is one critical transcription factor required for maximal expression of many cytokines involved in the pathogenesis of acute lung injury. Activation and regulation of NF-kappa B are tightly controlled by a complicated signaling cascade. In acute lung injury caused by infection of bacteria, Toll-like receptors play a central role in initiating the innate immune system and activating NF-kappa B. Anti-inflammatory cytokines such as interleukin-10 and interleukin-13 have been shown to suppress inflammatory processes through inhibiting NF-kappa B activation. NF-kappa B can interact with other transcription factors, and these interactions thereby lead to greater transcriptional selectivity. Modification of transcription is likely to be a logical therapeutic target for acute lung injury.


Asunto(s)
Proteínas de Drosophila , Pulmón/metabolismo , FN-kappa B/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Transducción de Señal , Transcripción Genética , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/fisiopatología , Fenómenos Fisiológicos Bacterianos , Regulación de la Expresión Génica/fisiología , Humanos , Pulmón/patología , Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , FN-kappa B/genética , Receptores de Superficie Celular/metabolismo , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/inmunología , Transducción de Señal/genética , Receptores Toll-Like , Factores de Transcripción/metabolismo
6.
J Biol Chem ; 276(44): 40977-81, 2001 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-11533055

RESUMEN

Chemoattractants bind to seven transmembrane-spanning, G-protein-coupled receptors on monocytes and neutrophils and induce a variety of functional responses, including activation of the transcription factor NF-kappaB. The signaling mechanisms utilized by chemoattractants to activate NF-kappaB in human peripheral blood monocytes are poorly defined. We previously demonstrated that fMet-Leu-Phe (fMLP) stimulates NF-kappaB activation, and this function of fMLP requires phosphatidylinositol 3-kinase (PI3K). Here we present evidence that fMLP activates RhoA and that fMLP-induced NF-kappaB activation requires this small GTPase. Stimulation of monocytes with fMLP rapidly activated RhoA as well as NF-kappaB, and their activation was markedly reduced by pertussis toxin treatment. Pretreatment of monocyte with a RhoA inhibitor, C3 transferase from Clostridium botulinum, effectively blocked fMLP-induced NF-kappaB activation as well as interleukin-1beta gene expression. A dominant negative form of RhoA (T19N) also inhibited fMLP-stimulated reporter gene expression in a kappaB-dependent manner. Cotransfection of the monocytic THP1 cells with a constitutively active form of RhoA (Q63L) with the promoter reporter plasmid results in a marked increase in NF-kappaB-mediated reporter gene expression. Furthermore, the PI3K inhibitors wortmannin and LY294002 block RhoA activation induced by fMLP. These results demonstrate that low molecular weight GTPase RhoA is a novel signal transducer for fMLP-induced NF-kappaB activation and Galpha(i) or Galpha(o) class of heterotrimeric G proteins likely mediate RhoA activation via PI3K in human peripheral blood monocytes.


Asunto(s)
N-Formilmetionina Leucil-Fenilalanina/farmacología , FN-kappa B/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Secuencia de Bases , Cloranfenicol O-Acetiltransferasa/genética , Cartilla de ADN , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Peso Molecular , Toxina del Pertussis , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Formil Péptido , Receptores Inmunológicos/metabolismo , Receptores de Péptidos/metabolismo , Transducción de Señal , Factores de Virulencia de Bordetella/metabolismo , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/química
7.
Mol Cell Biol ; 21(16): 5554-65, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11463837

RESUMEN

The procoagulant thrombin promotes the adhesion of polymorphonuclear leukocytes to endothelial cells by a mechanism involving expression of intercellular adhesion molecule 1 (ICAM-1) via an NF-kappaB-dependent pathway. We now provide evidence that protein kinase C-delta (PKC-delta) and the p38 mitogen-activated protein (MAP) kinase pathway play a critical role in the mechanism of thrombin-induced ICAM-1 gene expression in endothelial cells. We observed the phosphorylation of PKC-delta and p38 MAP kinase within 1 min after thrombin challenge of human umbilical vein endothelial cells. Pretreatment of these cells with the PKC-delta inhibitor rottlerin prevented the thrombin-induced phosphorylation of p38 MAP kinase, suggesting that p38 MAP kinase signals downstream of PKC-delta. Inhibition of PKC-delta or p38 MAP kinase by pharmacological and genetic approaches markedly decreased the thrombin-induced NF-kappaB activity and resultant ICAM-1 expression. The effects of PKC-delta inhibition were secondary to inhibition of IKKbeta activation and of subsequent NF-kappaB binding to the ICAM-1 promoter. The effects of p38 MAP kinase inhibition occurred downstream of IkappaBalpha degradation without affecting the DNA binding function of nuclear NF-kappaB. Thus, PKC-delta signals thrombin-induced ICAM-1 gene transcription by a dual mechanism involving activation of IKKbeta, which mediates NF-kappaB binding to the ICAM-1 promoter, and p38 MAP kinase, which enhances transactivation potential of the bound NF-kappaB p65 (RelA).


Asunto(s)
Endotelio Vascular/fisiología , Molécula 1 de Adhesión Intercelular/genética , Isoenzimas/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteína Quinasa C/fisiología , Trombina/fisiología , Células Cultivadas , Activación Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , FN-kappa B/fisiología , Proteína Quinasa C-delta , Transducción de Señal/efectos de los fármacos , Trombina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
8.
J Biol Chem ; 276(24): 20827-30, 2001 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-11331270

RESUMEN

Protein kinase A (PKA) is an important effector enzyme commonly activated by cAMP. The present study focuses on our finding that the vasoactive peptide endothelin-1 (ET1), whose signaling is not coupled to cAMP production, stimulates PKA in two independent cellular models. Using an in vivo assay for PKA activity, we found that ET1 stimulated PKA in HeLa cells overexpressing ET1 receptors and in aortic smooth muscle cells expressing endogenous levels of ET1 receptors. In these cell models, ET1 did not stimulate cAMP production, indicating a novel mechanism for PKA activation. The ET1-induced activation of PKA was found to be dependent on the degradation of inhibitor of kappaB, which was previously reported to bind and inhibit PKA. ET1 potently stimulated the nuclear factor-kappaB pathway, and this effect was inhibited by overexpression of the inhibitor of kappaB dominant negative mutant (IkappaBalpham) and by treatment with the proteasome inhibitor MG-132. Importantly, IkappaBalpham and MG-132 had similar inhibitory effects on ET1-induced activation of PKA without affecting G(s)-mediated activation of PKA or ET1-induced phosphorylation of mitogen-activated protein kinase. Finally, another vasoactive peptide, angiotensin II, also stimulated PKA in a cAMP-independent manner in aortic smooth muscle cells. These findings suggest that cAMP-independent activation of PKA might be a general response to vasoactive peptides.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Endotelina-1/farmacología , Proteínas I-kappa B , Isoproterenol/farmacología , Receptores de Endotelina/fisiología , Animales , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Células HeLa , Humanos , Leupeptinas/farmacología , Ratones , Complejos Multienzimáticos/metabolismo , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal , Receptor de Endotelina A , Receptores de Endotelina/genética , Proteínas Recombinantes/metabolismo , Transfección
9.
J Immunol ; 166(11): 6885-92, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11359849

RESUMEN

The guanine nucleotide-binding regulatory protein alpha-subunit, Galpha(16), is primarily expressed in hemopoietic cells, and interacts with a large number of seven-membrane span receptors including chemoattractant receptors. We investigated the biological functions resulting from Galpha(16) coupling of chemoattractant receptors in a transfected cell model system. HeLa cells expressing a kappaB-driven luciferase reporter, Galpha(16), and the formyl peptide receptor responded to fMLP with a approximately 7- to 10-fold increase in luciferase activity. This response was accompanied by phosphorylation of IkappaBalpha and elevation of nuclear kappaB-DNA binding activity, indicating activation of NF-kappaB. In contrast to Galpha(16), expression of Galpha(q), Galpha(13), and Galpha(i2) resulted in a marginal increase in kappaB luciferase activity. A GTPase-deficient, constitutively active Galpha(16) mutant (Q212L) could replace agonist stimulation for activation of NF-kappaB. Furthermore, expression of Galpha(16) (Q212L) markedly enhanced TNF-alpha-induced kappaB reporter activity. The Galpha(16)-mediated NF-kappaB activation was paralleled by an increase in phospholipase C-beta activity, and was blocked by pharmacological inhibitors of protein kinase C (PKC) and by buffering of intracellular Ca(2+). The involvement of a conventional PKC isoform was confirmed by the finding that expression of PKCalpha enhanced the effect of Galpha(16), and a dominant negative PKCalpha partially blocked Galpha(16)-mediated NF-kappaB activation. In addition to formyl peptide receptor, Galpha(16) also enhanced NF-kappaB activation by the C5a and C3a receptors, and by CXC chemokine receptor 2 and CCR8. These results suggest a potential role of Galpha(16) in transcriptional regulation downstream of chemoattractant receptors.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/fisiología , FN-kappa B/metabolismo , Receptores de Quimiocina/metabolismo , Transporte Activo de Núcleo Celular/genética , Sustitución de Aminoácidos/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Activación Enzimática/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Genes Reporteros , Glutamina/genética , Células HeLa , Proteínas de Unión al GTP Heterotriméricas/biosíntesis , Proteínas de Unión al GTP Heterotriméricas/genética , Humanos , Isoenzimas/metabolismo , Isoenzimas/fisiología , Leucina/genética , Luciferasas/genética , Fosfolipasa C beta , Receptores de Quimiocina/biosíntesis , Receptores de Quimiocina/genética , Receptores de Formil Péptido , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/genética , Receptores de Péptidos/biosíntesis , Receptores de Péptidos/genética , Transducción de Señal/genética , Transactivadores/genética , Transactivadores/fisiología , Transfección/métodos , Fosfolipasas de Tipo C/metabolismo , Fosfolipasas de Tipo C/fisiología
10.
J Immunol ; 166(6): 4099-105, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11238659

RESUMEN

The NPXXY motif (X represents any amino acid) in the seventh transmembrane domain of the chemotactic formyl peptide receptor (FPR) is highly conserved among G protein-coupled receptors. Recent work suggested that this motif contributes to G protein-coupled receptor internalization and signal transduction; however, its role in FPR signaling remains unclear. In this study we replaced Asn(297) and Tyr(301) in the NPXXY motif of the human FPR with Ala (N297A) and Ala/Phe (Y301A/Y301F), respectively, and determined the effects of the substitutions on FPR functions in transfected rat basophilic leukemia cells. Whereas all the mutant receptors were expressed on the cell surface, the N297A receptor exhibited reduced binding affinity and was unable to mediate activation of phospholipase C-beta and the p42/44 mitogen-activated protein kinase (MAP kinase). The Y301F receptor displayed significantly decreased ligand-stimulated internalization and MAP kinase activation, suggesting that the hydrogen bonding at Tyr(301) is critical for these functions. The Y301F receptor showed a chemotactic response similar to that of wild-type FPR, indicating that cell chemotaxis does not require receptor internalization and hydrogen bonding at the Tyr(301) position. In contrast, the Y301A receptor displayed a left-shifted, but overall reduced, chemotaxis response that peaked at 0.1-1 nM. Finally, using a specific MAP kinase kinase inhibitor, we found that activation of MAP kinase is required for efficient FPR internalization, but is not essential for chemotaxis. These findings demonstrate that residues within the NPXXY motif differentially regulate the functions of FPR.


Asunto(s)
N-Formilmetionina Leucil-Fenilalanina/metabolismo , Fragmentos de Péptidos/inmunología , Receptores Inmunológicos/fisiología , Receptores de Péptidos/fisiología , Transducción de Señal/inmunología , Secuencias de Aminoácidos/genética , Secuencias de Aminoácidos/inmunología , Animales , Butadienos/farmacología , Calcio/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Quimiotaxis de Leucocito/inmunología , Secuencia Conservada/genética , Secuencia Conservada/inmunología , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutagénesis Sitio-Dirigida , Nitrilos/farmacología , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Unión Proteica/genética , Unión Proteica/inmunología , Ratas , Receptores de Formil Péptido , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores de Péptidos/biosíntesis , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Transducción de Señal/genética , Células Tumorales Cultivadas
11.
J Biol Chem ; 276(16): 13039-48, 2001 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-11278473

RESUMEN

The cGMP-dependent protein kinases (PKGs) are ubiquitous effector enzymes that regulate a variety of physiological processes in response to nitric oxide and natriuretic agonists. We have constructed green fluorescent fusion proteins (GFP) using full-length (PKG-GFP) and truncations encoding either the regulatory domain of PKG1alpha (G1alphaR-GFP) or the catalytic domains of PKG1alpha (GFP-G1C) to examine the enzymatic properties and intracellular location. When transiently transfected into mammalian cells, these constructs were detected on Western blots at the expected sizes using anti-GFP antibodies. The GFP-G1C and the full-length PKG1alpha-GFP fusion proteins were found to have constitutive activity both in vivo and in vitro. The G1alphaR-GFP protein was found to dimerize with endogenous type 1 PKG and behaved in a dominant negative manner both in vivo and in vitro. When expressed transiently in either HEK-293 or A549 epithelial cells, the fusion proteins encoding the amino-terminal regulatory domains (PKG-GFP, G1alphaR-GFP) were present in the cytosol and were rarely observed in the nucleus. In contrast, the GFP-G1C (lacking regulatory domains) concentrated in the nucleus. Of the fusion proteins containing the regulatory region, the constitutive PKG-GFP protein was present in a more centralized location, whereas the G1alphaR-GFP protein colocalized with F-actin on stress fibers and in dynamic regions of the plasma membrane. Microscopic and immunoprecipitation studies indicated that both the G1alphaR-GFP and the PKG-GFP fusion proteins colocalized with vasodilator-stimulated phosphoprotein (VASP). These constructs thus represent novel tools with which to visualize inactive, and activated, PKG1alpha in vivo, and we have used them to demonstrate two functionally independent domains. In addition, we show for the first time in living cells that PKG is found in dynamic membrane regions in association with VASP.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Actinas/análisis , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Línea Celular , Membrana Celular/enzimología , Núcleo Celular/enzimología , Proteínas Quinasas Dependientes de GMP Cíclico/análisis , Proteínas Quinasas Dependientes de GMP Cíclico/química , Proteínas Fluorescentes Verdes , Humanos , Riñón , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Mucosa Respiratoria , Transfección
12.
Physiol Genomics ; 4(2): 137-145, 2000 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-11120874

RESUMEN

We investigated the function of proteinase-activated receptor-1 (PAR-1) in the regulation of pulmonary microvascular permeability in response to thrombin challenge using PAR-1 knockout mice (-/-). Lungs were isolated and perfused with albumin (5 g/100 ml)-Krebs solution at constant flow (2 ml/min). Lung wet weight and pulmonary artery pressure (P(pa)) were continuously monitored. We determined the capillary filtration coefficient (K(fc)) and (125)I-labeled albumin (BSA) permeability-surface area product (PS) to assess changes in pulmonary microvessel permeability to liquid and albumin, respectively. Normal and PAR-1-null lung preparations received in the perfusate: 1) thrombin or 2) selective PAR-1 agonist peptide (TFLLRNPNDK-NH(2)). In control PAR-1 (+/+) mouse lungs, (125)I-albumin PS and K(fc) were significantly increased over baseline (by approximately 7- and 1.5-fold, respectively) within 20 min of alpha-thrombin (100 nM) challenge. PAR-1 agonist peptide (5 microM) gave similar results, whereas control peptide (5 microM; FTLLRNPNDK-NH(2)) was ineffective. At relatively high concentrations, thrombin (500 nM) or PAR-1 agonist peptide (10 microM) also induced increases in P(pa) and lung wet weight. All effects of thrombin (100 or 500 nM) or PAR-1 agonist peptide (5 or 10 microM) were prevented in PAR-1-null lung preparations. Baseline measures of microvessel permeability and P(pa) in the PAR-1-null preparations were indistinguishable from those in normal lungs. Moreover, PAR-1-null preparations gave normal vasoconstrictor response to thromboxane analog, U-46619 (100 nM). The results indicate that the PAR-1 receptor is critical in mediating the permeability-increasing and vasoconstrictor effects of thrombin in pulmonary microvessels.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Circulación Pulmonar/efectos de los fármacos , Receptores de Trombina/genética , Trombina/farmacología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Relación Dosis-Respuesta a Droga , Genotipo , Técnicas In Vitro , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Ratones , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Oligopéptidos/farmacología , Fosforilación/efectos de los fármacos , ARN/genética , ARN/metabolismo , Receptor PAR-1 , Receptores de Trombina/fisiología , Vasoconstrictores/farmacología
13.
Am J Physiol Lung Cell Mol Physiol ; 279(6): L1129-36, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11076803

RESUMEN

Interleukin (IL)-8 is a C-X-C chemokine that plays an important role in acute inflammation through its G protein-coupled receptors CXCR1 and CXCR2. In this study, we investigated the role of IL-8 as an autocrine regulator of IL-8 production and the signaling mechanisms involved in human peripheral blood mononuclear cells (MNCs). Sepharose-immobilized IL-8 stimulated a sevenfold increase in IL-8 production within 2 h. IL-8 induced the expression of its own message, and IL-8 biosynthesis was inhibited by cycloheximide and actinomycin D, indicating de novo RNA and protein synthesis. In contrast to MNCs, polymorphonuclear neutrophils did not respond to the immobilized IL-8 with IL-8 production despite cell surface expression of CXCR1 and CXCR2. Melanoma growth-stimulatory activity/growth-related protein-alpha (MGSA/GROalpha), which binds CXCR2 but not CXCR1, was unable to either stimulate IL-8 secretion in MNCs or desensitize these cells to respond to immobilized IL-8. The involvement of mitogen-activated protein kinase (MAPK) in IL-8-induced IL-8 biosynthesis was suggested by the ability of PD-98059, an inhibitor of MAPK kinase, to block this function. Furthermore, IL-8 induced a significant increase in extracellular signal-regulated kinase 2 phosphorylation, whereas MGSA/GROalpha was much less effective. These findings support the role of IL-8 as an autocrine regulator of IL-8 production and suggest that this function is mediated by CXCR1 through activation of MAPK.


Asunto(s)
Comunicación Autocrina/inmunología , Quimiocinas CXC , Péptidos y Proteínas de Señalización Intercelular , Interleucina-8/metabolismo , Monocitos/enzimología , Monocitos/inmunología , Anticuerpos Monoclonales , Quimiocina CXCL1 , Factores Quimiotácticos/farmacología , Cicloheximida/farmacología , Dactinomicina/farmacología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Citometría de Flujo , Inhibidores de Crecimiento/farmacología , Sustancias de Crecimiento/farmacología , Humanos , Interleucina-8/genética , Interleucina-8/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Monocitos/química , Neumonía/inmunología , Neumonía/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/inmunología , Inhibidores de la Síntesis de la Proteína/farmacología , Receptores de Interleucina-8A/análisis , Receptores de Interleucina-8A/inmunología , Receptores de Interleucina-8B/análisis , Receptores de Interleucina-8B/inmunología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/inmunología
14.
Gene ; 257(2): 307-17, 2000 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-11080597

RESUMEN

This study delineates the isolation and characterization of a novel chemokine receptor gene, the putative chicken CXC receptor 1 (cCXCR1). Using a human CXCR1 probe, we isolated several positive clones from a chicken genomic library. One of the clones contained a fragment of approximately 5000bp that hybridized strongly with the hCXCR1 probe. This fragment was sequenced and subjected to a variety of computer analyses. The open reading frame for this gene predicts a seven transmembrane domain protein with all the characteristics of a chemokine receptor and with 67% sequence homology to hCXCR1, 65% to hCXCR2 and also with considerable sequence homology to other human chemokine receptors such as hCXCR4 (50%), hCCR2 (49%) and hCCR1 (49%). However, the homology to a previously isolated potential G-protein-coupled receptor for chickens (AvCRL1) is only 47%. Using 5' RACE, two transcription initiation sites were identified suggesting the potential for the expression of two protein isoforms (I and II) in vivo. The promoter for the putative cCXCR1 contains a variety of consensus transcription factor binding elements that can potentially be involved in the expression of this chicken receptor upon stimulation by stress-inducing agents. RT-PCR analysis was used to determine the pattern of expression of the larger isoform (I) of this receptor in a variety of tissues. This form of the receptor is expressed primarily in the organs of the gastrointestinal tract, tissues that are frequently exposed to stress-inducing agents, but not in the central nervous system, tissues that are protected from insult by the blood barrier. Using the same RT-PCR approach we show that stress-inducing agents, such as 'first-hand' and 'second-hand' cigarette smoke components, tumor promoters and thrombin, differentially stimulate the expression of the isoform I in primary fibroblasts. Thrombin is an enzyme that plays many important roles in thrombosis, angiogenesis and wound healing and exposure to both cigarette smokes and/or to tumor promoters can lead to tumorigenesis. Therefore, upregulation of chemokines and their receptors by stress-inducing agents can confer highly regulated modulation of cellular responses to traumatic and pathological situations.


Asunto(s)
Genes/genética , Receptores de Quimiocina/genética , Receptores de Interleucina-8A/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Cultivadas , Pollos , Clonación Molecular , ADN/química , ADN/genética , ADN/aislamiento & purificación , Regulación de la Expresión Génica/efectos de los fármacos , Datos de Secuencia Molecular , Ésteres del Forbol/farmacología , Isoformas de Proteínas/genética , ARN/genética , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Trombina/farmacología , Distribución Tisular , Transcripción Genética
15.
J Immunol ; 165(8): 4598-605, 2000 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11035102

RESUMEN

Formyl peptides are potent neutrophil chemoattractants. In humans and rabbits, the formyl peptide receptor (FPR) binds N-formyl-Met-Leu-Phe (fMLF) with high affinity (K(d) approximately 1 nM). The mouse FPR (mFPR) is a low-affinity receptor for fMLF (K(d) approximately 100 nM); therefore, other agonists for this receptor may exist. Using mFPR-transfected rat basophilic leukemia cells, we found that a recently identified synthetic peptide Trp-Lys-Tyr-Met-Val-D-Met (WKYMVm) is a potent agonist for mFPR. WKYMVm induced calcium mobilization with an EC(50) of 1.2-1.5 nM. Optimal chemotaxis was achieved with 1 nM of WKYMVm, but it required 100 nM of fMLF. WKYMVm stimulated rapid and potent phosphorylation of the mitogen-activated protein kinases extracellular signal-related kinases 1 and 2 when used at 50 nM. Pertussis toxin only partially blocked calcium mobilization and production of inositol 1,4,5-trisphosphate in the stimulated mFPR cells, suggesting the possibility that this receptor couples to Galpha proteins other than Gi and Go. Competitive binding and desensitization data suggest that both peptides interact with the same receptor but may use nonoverlapping binding sites because WKYMVm was unable to effectively displace [(3)H]fMLF bound to mFPR. These results provide evidence for the presence of an alternative potent agonist for mFPR, and suggest a potential usage of WKYMVm for probing the ligand-receptor interactions with the murine formyl peptide receptor homologs.


Asunto(s)
Factores Quimiotácticos/inmunología , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Oligopéptidos/agonistas , Oligopéptidos/inmunología , Receptores Inmunológicos/agonistas , Receptores Inmunológicos/inmunología , Receptores de Péptidos/agonistas , Receptores de Péptidos/inmunología , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/inmunología , Quimiotaxis/inmunología , Activación Enzimática/inmunología , Espacio Extracelular/inmunología , Espacio Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Oligopéptidos/metabolismo , Toxina del Pertussis , Unión Proteica/inmunología , Ratas , Receptores de Formil Péptido , Receptores Inmunológicos/metabolismo , Receptores de Péptidos/metabolismo , Células Tumorales Cultivadas , Factores de Virulencia de Bordetella/toxicidad
17.
J Biol Chem ; 275(32): 24907-14, 2000 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-10801799

RESUMEN

Recent work has suggested a role for the serine/threonine kinase Akt and IkappaB kinases (IKKs) in nuclear factor (NF)-kappaB activation. In this study, the involvement of these components in NF-kappaB activation through a G protein-coupled pathway was examined using transfected HeLa cells that express the B2-type bradykinin (BK) receptor. The function of IKK2, and to a lesser extent, IKK1, was suggested by BK-induced activation of their kinase activities and by the ability of their dominant negative mutants to inhibit BK-induced NF-kappaB activation. BK-induced NF-kappaB activation and IKK2 activity were markedly inhibited by RGS3T, a regulator of G protein signaling that inhibits Galpha(q), and by two Gbetagamma scavengers. Co-expression of Galpha(q) potentiated BK-induced NF-kappaB activation, whereas co-expression of either an activated Galpha(q)(Q209L) or Gbeta(1)gamma(2) induced IKK2 activity and NF-kappaB activation without BK stimulation. BK-induced NF-kappaB activation was partially blocked by LY294002 and by a dominant negative mutant of phosphoinositide 3-kinase (PI3K), suggesting that PI3K is a downstream effector of Galpha(q) and Gbeta(1)gamma(2) for NF-kappaB activation. Furthermore, BK could activate the PI3K downstream kinase Akt, whereas a catalytically inactive mutant of Akt inhibited BK-induced NF-kappaB activation. Taken together, these findings suggest that BK utilizes a signaling pathway that involves Galpha(q), Gbeta(1)gamma(2), PI3K, Akt, and IKK for NF-kappaB activation.


Asunto(s)
Bradiquinina/farmacología , Subunidades beta de la Proteína de Unión al GTP , Subunidades gamma de la Proteína de Unión al GTP , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP Heterotriméricas , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Sustitución de Aminoácidos , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Proteínas de Unión al GTP/genética , Genes Reporteros , Células HeLa , Humanos , Quinasa I-kappa B , Interleucina-1/farmacología , Luciferasas/genética , Mutagénesis Sitio-Dirigida , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptor de Bradiquinina B2 , Receptores de Bradiquinina/genética , Receptores de Bradiquinina/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/farmacología
18.
J Leukoc Biol ; 67(4): 577-84, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10770292

RESUMEN

Fractalkine is an endothelial cell-derived CX3C chemokine that is chemotactic mainly to mononuclear cells. Fractalkine was induced in rat aortic endothelial cells (RAEC) by interleukin-1beta (IL-1beta), tumor necrosis factor alpha (TNF-alpha), and lipopolysaccharide (LPS) transcriptionally and translationally. This induction correlated with increased NF-kappaB DNA binding activity as determined by gel mobility shift assay. Supershift assays revealed that the NF-kappaB subunits p50 and p65 were responsible for kappaB binding. Accordingly, we examined the role of NF-kappaB in fractalkine induction in RAEC through the use of an adenovirus-mediated mutant IkappaB as a specific inhibitor. Delivery of a dominant-negative form of IkappaBalpha in RAEC dramatically reduced the induction of fractalkine by these stimuli, suggesting a role for NF-kappaB activation in fractalkine induction. The inhibition of fractalkine expression by two potent NF-kappaB inhibitors, sulfasalazine and sanguinarine, further supported the central role of NF-kappaB in fractalkine transcription regulation and suggested a novel therapeutic target aimed at modulating leukocyte endothelial cell interaction.


Asunto(s)
Quimiocinas CX3C , Quimiocinas CXC/biosíntesis , Endotelio Vascular/metabolismo , Interleucina-1/farmacología , Lipopolisacáridos/farmacología , Proteínas de la Membrana/biosíntesis , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Aorta , Células Cultivadas , Quimiocina CX3CL1 , Ratas , Regulación hacia Arriba/efectos de los fármacos
19.
J Cell Physiol ; 183(2): 254-64, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10737901

RESUMEN

Tumor cell migration may favor local mass expansion and metastasis dissemination. Several tumors were found to express the receptor for platelet-activating factor (PAF), a potent mediator of leukocyte chemotaxis and endothelial cell migration. However, its functional role on tumor cells is largely unexplored. In the present study, we evaluated the motogenic effect of PAF on Chinese hamster ovarian (CHO) cancer cells transfected with the human PAF-receptor cDNA (CHO PAF-R). By using time-lapse recording, we detected a rapid motogenic response to PAF stimulation on CHO PAF-R, whereas no effect was evident on vector-only transfected cells. Such an effect was observed on scattered cell motility, on cells seeded on a fibronectin- or collagen-coated surface, and on migration of confluent monolayer cells. Cell speed increased at 1 h and was maximal 6-8 h after PAF stimulation on CHO PAF-R. Concomitantly, PAF induced marked changes in cytoskeleton actin distribution with cell contraction, assembling of stress fibers, and polar foci of adhesion. In conclusion, the present study demonstrates that PAF is a potent inducer of tumor cell motility, thus suggesting a role for this mediator in tumor growth and dissemination.


Asunto(s)
Movimiento Celular/fisiología , Glicoproteínas de Membrana Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/fisiología , Receptores de Superficie Celular , Receptores Acoplados a Proteínas G , Animales , Secuencia de Bases , Células CHO , Movimiento Celular/efectos de los fármacos , Cricetinae , Cartilla de ADN/genética , Expresión Génica , Humanos , Microscopía por Video , Factor de Activación Plaquetaria/farmacología , Factor de Activación Plaquetaria/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección
20.
J Biol Chem ; 275(4): 2811-6, 2000 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-10644746

RESUMEN

An increase in cellular levels of cyclic nucleotides activates serine/threonine-dependent kinases that lead to diverse physiological effects. Recently we reported the activation of the p38 mitogen-activated protein kinase (MAPK) pathway in neutrophils by a cGMP-dependent mechanism. In this study we demonstrated that exogenously supplied nitric oxide leads to activation of p38 MAPK in 293T fibroblasts. Phosphorylation of p38 corresponded with an increase in ATF-2-dependent gene expression. The effect of nitric oxide was mimicked by addition of 8-bromo-cGMP, indicating that activation of soluble guanylyl cyclase was involved. The importance of cGMP-dependent protein kinase in the activation of p38 MAPK by nitric oxide in 293T cells was assessed in a transfection based assay. Overexpression of cGMP-dependent protein kinase-1alpha caused phosphorylation of p38 in these cells and potentiated the effectiveness of cGMP. Overexpression of a catalytically inactive mutant form of this enzyme (T516A) blocked the ability of both nitric oxide and 8-bromo-cGMP to activate p38 as measured by both p38 phosphorylation and ATF-2 driven gene expression. Together, these data demonstrate that nitric oxide stimulates a novel pathway leading to activation of p38 MAPK that requires activation of cGMP-dependent protein kinase.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Western Blotting , Línea Celular , Activación Enzimática , Fibroblastos/enzimología , Humanos , Óxido Nítrico , Fosforilación , Pruebas de Precipitina , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA