Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Anesthesiology ; 130(1): 41-54, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30550426

RESUMO

BACKGROUND: Although dantrolene effectively treats malignant hyperthermia (MH), discrepant recommendations exist concerning dantrolene availability. Whereas Malignant Hyperthermia Association of the United States guidelines state dantrolene must be available within 10 min of the decision to treat MH wherever volatile anesthetics or succinylcholine are administered, a Society for Ambulatory Anesthesia protocol permits Class B ambulatory facilities to stock succinylcholine for airway rescue without dantrolene. The authors investigated (1) succinylcholine use rates, including for airway rescue, in anesthetizing/sedating locations; (2) whether succinylcholine without volatile anesthetics triggers MH warranting dantrolene; and (3) the relationship between dantrolene administration and MH morbidity/mortality. METHODS: The authors performed focused analyses of the Multicenter Perioperative Outcomes Group (2005 through 2016), North American MH Registry (2013 through 2016), and Anesthesia Closed Claims Project (1970 through 2014) databases, as well as a systematic literature review (1987 through 2017). The authors used difficult mask ventilation (grades III and IV) as a surrogate for airway rescue. MH experts judged dantrolene treatment. For MH morbidity/mortality analyses, the authors included U.S. and Canadian cases that were fulminant or scored 20 or higher on the clinical grading scale and in which volatile anesthetics or succinylcholine were given. RESULTS: Among 6,368,356 queried outcomes cases, 246,904 (3.9%) received succinylcholine without volatile agents. Succinylcholine was used in 46% (n = 710) of grade IV mask ventilation cases (median dose, 100 mg, 1.2 mg/kg). Succinylcholine without volatile anesthetics triggered 24 MH cases, 13 requiring dantrolene. Among 310 anesthetic-triggered MH cases, morbidity was 20 to 37%. Treatment delay increased complications every 10 min, reaching 100% with a 50-min delay. Overall mortality was 1 to 10%; 15 U.S. patients died, including 4 after anesthetics in freestanding facilities. CONCLUSIONS: Providers use succinylcholine commonly, including during difficult mask ventilation. Succinylcholine administered without volatile anesthetics may trigger MH events requiring dantrolene. Delayed dantrolene treatment increases the likelihood of MH complications. The data reported herein support stocking dantrolene wherever succinylcholine or volatile anesthetics may be used.


Assuntos
Dantroleno/uso terapêutico , Hipertermia Maligna/tratamento farmacológico , Hipertermia Maligna/etiologia , Relaxantes Musculares Centrais/uso terapêutico , Fármacos Neuromusculares Despolarizantes/efeitos adversos , Succinilcolina/efeitos adversos , Bases de Dados Factuais , Humanos
2.
J Immunol ; 181(2): 1429-37, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18606697

RESUMO

The prototypic formyl peptide N-formyl-Met-Leu-Phe (fMLF) is a major chemoattractant found in Escherichia coli culture supernatants and a potent agonist at human formyl peptide receptor (FPR) 1. Consistent with this, fMLF induces bactericidal functions in human neutrophils at nanomolar concentrations. However, it is a much less potent agonist for mouse FPR (mFPR) 1 and mouse neutrophils, requiring micromolar concentrations for cell activation. To determine whether other bacteria produce more potent agonists for mFPR1, we examined formyl peptides from Listeria monocytogenes and Staphylococcus aureus for their abilities to activate mouse neutrophils. A pentapeptide (N-formyl-Met-Ile-Val-Ile-Leu (fMIVIL)) from L. monocytogenes and a tetrapeptide (N-formyl-Met-Ile-Phe-Leu (fMIFL)) from S. aureus were found to induce mouse neutrophil chemotaxis at 1-10 nM and superoxide production at 10-100 nM, similar to the potency of fMLF on human neutrophils. Using transfected cell lines expressing mFPR1 and mFPR2, which are major forms of FPRs in mouse neutrophils, we found that mFPR1 is responsible for the high potency of fMIVIL and fMIFL. In comparison, activation of mFPR2 requires micromolar concentrations of the two peptides. Genetic deletion of mfpr1 resulted in abrogation of neutrophil superoxide production and degranulation in response to fMIVIL and fMIFL, further demonstrating that mFPR1 is the primary receptor for detection of these formyl peptides. In conclusion, the formyl peptides from L. monocytogenes and S. aureus are approximately 100-fold more potent than fMLF in activating mouse neutrophils. The ability of mFPR1 to detect bacterially derived formyl peptides indicates that this important host defense mechanism is conserved in mice.


Assuntos
Fatores Quimiotáticos/imunologia , Quimiotaxia de Leucócito , Listeria monocytogenes/imunologia , Neutrófilos/imunologia , Oligopeptídeos/imunologia , Receptores de Formil Peptídeo/metabolismo , Staphylococcus aureus/imunologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Formilmetionina Leucil-Fenilalanina/imunologia , Ativação de Neutrófilo , Neutrófilos/metabolismo , Oligopeptídeos/metabolismo , Ratos , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/imunologia , Transfecção
3.
Methods Enzymol ; 389: 229-43, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15313569

RESUMO

Regulator of G-protein signaling (RGS) proteins are very active GTPase-accelerating proteins (GAPs) in vitro and are expected to reduce signaling by G-protein coupled receptors in vivo. A novel method is presented to assess the in vivo role of RGS proteins in the function of a G protein in which Galpha subunits do not bind to RGS proteins or respond with enhanced GTPase activity. A point mutation in the switch I region of Galpha subunits (G184S Galpha(o) and G183S Galpha(i1)) blocks the interaction with RGS proteins but leaves intact the ability of Galpha to couple to betagamma subunits, receptors, and downstream effectors. Expression of the RGS-insensitive mutant G184S Galpha(o) in C6 glioma cells with the micro-opioid receptor dramatically enhances adenylylcyclase inhibition and activation of extracellular regulated kinase. Introducing the same G184S Galpha(o) protein into embryonic stem (ES) cells by gene targeting allows us to assess the functional importance of the endogenous RGS proteins using in vitro differentiation models and in intact mice. Using ES cell-derived cardiocytes, spontaneous and isoproterenol-stimulated beating rates were not different between wild-type and G184S Galpha(o) mutant cells; however, the bradycardiac response to adenosine A1 receptor agonists was enhanced significantly (seven-fold decrease EC50) in Galpha(o)RGSi mutant cells compared to wild-type Galpha(o), indicating a significant role of endogenous RGS proteins in cardiac automaticity regulation. The approach of using RGS-insensitive Galpha subunit knockins will reveal the role of RGS protein-mediated GAP activity in signaling by a given G(i/o) protein. This will reveal the full extent of RGS regulation and will not be confounded by redundancy in the function of multiple RGS proteins.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/genética , Mutação Puntual , Proteínas RGS/metabolismo , Transdução de Sinais , Animais , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Proteínas RGS/genética
4.
J Immunol ; 178(1): 416-27, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17182580

RESUMO

We examined the roles of cGMP-dependent protein kinase (PKG) and PI3K in degranulation induced by fMLF and by FcepsilonRI cross-linking. In rat basophilic leukemia-2H3 cells expressing formyl peptide receptor, the PKG inhibitors KT5823 and Rp-8-Br-PET-cGMP, as well as the PI3K inhibitor LY294002, reduced agonist-stimulated beta-hexosaminidase release in a dose-dependent manner. These inhibitors also abolished vesicular fusion with the plasma membrane, as evidenced by diminished annexin V staining. Agonist-induced degranulation was completely blocked when LY294002 was applied together with one of the PKG inhibitors, suggesting an additive and possibly synergistic effect. In contrast, the PKG inhibitors did not affect fMLF-induced intracellular calcium mobilization and Akt phosphorylation. Likewise, LY294002 did not alter fMLF-induced elevation of intracellular cGMP concentration, and the inhibitory effect of LY294002 was not reversed by a cell-permeable analog of cGMP. Treatment with fMLF induced phosphorylation of soluble N-ethylmaleimide-sensitive factor-attachment protein (SNAP)-23, syntaxins 2, 4, and 6, and Monc18-3. The induced phosphorylation of SNAP-23 and syntaxins 2 and 4 was blocked by Rp-8-Br-PET-cGMP and LY294002. However, LY294002 was less effective in inhibiting Munc18-3 phosphorylation. The induced phosphorylation of syntaxin 6 was not effectively blocked by either Rp-8-Br-PET-cGMP or LY294002. Treatment of human neutrophils with the PKG inhibitors and LY294002 reduced enzyme release from primary, secondary, and tertiary granules. These results suggest that PKG and PI3K are involved in degranulation, possibly through phosphorylation of target membrane SNAP receptor proteins and their binding proteins.


Assuntos
Degranulação Celular , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Neutrófilos/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas SNARE/metabolismo , Animais , Carbazóis/farmacologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Cromonas/farmacologia , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Humanos , Indóis/farmacologia , Leucócitos/efeitos dos fármacos , Leucócitos/enzimologia , Leucócitos/imunologia , Mastócitos/metabolismo , Proteínas de Membrana/metabolismo , Morfolinas/farmacologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos , Receptores de IgE/antagonistas & inibidores , Tionucleotídeos/farmacologia
5.
Mol Pharmacol ; 72(4): 976-83, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17652444

RESUMO

A series of quinazolinone derivatives were synthesized based on a hit compound identified from a high-throughput screening campaign targeting the human formyl peptide receptor-like 1 (FPRL1). Based on structure-activity relationship analysis, we found that substitution on the para position of the 2-phenyl group of the quinazolinone backbone could alter the pharmacological properties of the compound. The methoxyl substitution produced an agonist 4-butoxy-N-[2-(4-methoxy-phenyl)-4-oxo-1,4-dihydro-2H-quinazolin-3-yl]-benzamide (Quin-C1; C1), whereas a hydroxyl substitution resulted in a pure antagonist, Quin-C7 (C7). Several partial agonists were derived from other substitutions on the para position. C7 partially displaced [(125)I]Trp-Lys-Tyr-Met-Val-d-Met-NH(2) (WKYMVm) binding to FPRL1 but not [(3)H]N-formyl-Met-Leu-Phe to formyl peptide receptor. In functional assays using FPRL1-expressing RBL-2H3 cells, C7 inhibited calcium mobilization and chemotaxis induced by WKYMVm and C1 and degranulation elicited by C1. C7 also suppressed C1-induced extracellular signal-regulated kinase phosphorylation and reduced arachidonic acid-induced ear edema in mice. This study represents the first characterization of a nonpeptidic antagonist for FPRL1 and suggests the prospect of using low molecular weight compounds as modulators of chemoattractant receptors in vitro and in vivo.


Assuntos
Fatores Quimiotáticos/farmacologia , Oligopeptídeos/farmacologia , Receptores de Formil Peptídeo/antagonistas & inibidores , Ácido Araquidônico/farmacologia , Cálcio/metabolismo , Linhagem Celular , Quimiotaxia/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Humanos , Ligantes , Fosforilação , Receptores de Formil Peptídeo/genética
6.
J Immunol ; 177(10): 7050-8, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17082621

RESUMO

Cyclosporin A (CsA) is a fungus-derived cyclic undecapeptide with potent immunosuppressive activity. Its analog, cyclosporin H (CsH), lacks immunosuppressive function but can act as an antagonist for the human formyl peptide receptor (FPR). More recent studies have shown that CsA also inhibits fMLF-induced degranulation in differentiated HL-60 promyelocytic leukemia cells. However, it is unclear whether CsA interferes with ligand-receptor interaction, G protein activation, or other downstream signaling events. In this study we used human neutrophils, differentiated HL-60 cells, and rat basophilic leukemia (RBL)-2H3 cells expressing human FPR (RBL-FPR) to identify the action site of CsA. In functional assays, CsA inhibited fMLF-stimulated degranulation, chemotaxis, calcium mobilization, and phosphorylation of the MAPKs ERK 1/2 and the serine/threonine protein kinase Akt. CsA also blocked Trp-Lys-Tyr-Met-Val-D-Met (WKYMVm)-induced functions in RBL-FPR cells. Concentrations for half-maximal inhibition with CsA are generally 6- to 50-fold higher than that of CsH. CsA was compared with another immunosuppressant, ascomycin, relative to the inhibitory effects on FPR-mediated chemotaxis, calcium mobilization, and degranulation. In these experiments, ascomycin produced no inhibitory effects at low micromolar concentrations (1-4 microM), whereas the inhibitory effects of CsA were prominent at comparable concentrations. Finally, CsA dose-dependently inhibited the uptake of fNle-Leu-Phe-Nle-Tyr-Lys-fluoresceine and [3H]fMLF or [125I]WKYMVm binding to FPR. However, CsA and CsH did not show any obvious inhibitory effect on FPR-like 1-mediated cellular functions. These results demonstrate that CsA is a selective antagonist of FPR and that its inhibition of fMLF-stimulated leukocyte activation is at the level of cognate ligand binding.


Assuntos
Ciclosporina/farmacologia , Imunossupressores/farmacologia , Receptores de Formil Peptídeo/antagonistas & inibidores , Receptores de Formil Peptídeo/metabolismo , Animais , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/imunologia , Cálcio/antagonistas & inibidores , Cálcio/metabolismo , Degranulação Celular/efeitos dos fármacos , Degranulação Celular/imunologia , Linhagem Celular Tumoral , Inibição de Migração Celular , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Células HL-60 , Humanos , Ligantes , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/imunologia , Ratos , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia
7.
Immunol Invest ; 33(2): 193-212, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15195697

RESUMO

The N-formyl peptide receptor (FPR) is a G protein-coupled chemoattractant receptor that mediates diverse leukocyte functions when stimulated by bacteria-derived N-formyl peptides such as fMet-Leu-Phe (fMLF). Impaired neutrophil responsiveness to fMLF parallels increased susceptibility to periodontal microorganisms among patients with localized juvenile periodontitis (LJP). To determine whether the recently identified FPR mutations in LJP patients are responsible for selective loss of receptor-mediated functions, we prepared and analyzed RBL-2H3 cells expressing FPR bearing Phe110 to Ser (FPR-F110S) or Cys-126 to Trp (FPR-C126W) replacement as well as a FPR double mutant (FPR-FSCW). All mutant receptors were expressed normally on the cell surface, but were unable to mediate release of beta-hexosaminidase upon fMLF stimulation. FPR-C126W effectively mediated fMLF uptake, an indication of receptor-mediated endocytosis, whereas FPR-F110S and FSCW exhibited markedly reduced ability to uptake fMLF. Both FPR-F110S and FPR-C126W were defective in chemotaxis and displayed reduced Ca2+ mobilization, but mutation at both positions partially restored the ability to respond to fMLF in chemotaxis assay and was nearly normal in Ca2+ mobilization assay. All mutants exhibited diminished accumulation of inositol phosphates. FPR-F110S displayed a delayed and significantly reduced ERK phosphorylation whereas FPR-FSCW nearly lost the ability to phosphorylate ERK. Taken together, these results indicate compromised signaling capabilities due to the FPR mutations, but the loss of function is selective and could be partially rescued by mutations at both positions.


Assuntos
Substituição de Aminoácidos/genética , Membrana Celular/metabolismo , Regulação da Expressão Gênica , Receptores de Formil Peptídeo/química , Receptores de Formil Peptídeo/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Quimiotaxia/genética , Humanos , Isoenzimas/metabolismo , Ligantes , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Toxina Pertussis/farmacologia , Fosfolipase C beta , Fosforilação , Ratos , Receptores de Formil Peptídeo/genética , Relação Estrutura-Atividade , Fosfolipases Tipo C/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
8.
J Immunol ; 173(12): 7462-70, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15585872

RESUMO

A whole-cell-based reconstitution system was developed to study the signaling mechanisms underlying chemoattractant-induced activation of NADPH oxidase. This system takes advantage of the lack of formyl peptide receptor-mediated response in COS-phox cells expressing gp91(phox), p22(phox), p67(phox), and p47(phox), which respond to phorbol ester and arachidonic acid with O()(2) production. By exogenous expression of signaling molecules enriched in neutrophils, we have identified several critical components for fMLP-induced NADPH oxidase activation. Expression of PKCdelta, but not PKCalpha, -betaII, and -zeta, is necessary for the COS-phox cells to respond to fMLP. A role of PKCdelta in neutrophil NADPH oxidase was confirmed based on the ability of fMLP to induce PKCdelta translocation and the sensitivity of fMLP-induced O()(2) production to rottlerin, a PKCdelta-selective inhibitor. Optimal reconstitution also requires phospholipase C-beta2 and PI3K-gamma. We found that formyl peptide receptor could use the endogenous Rac1 as well as exogenous Rac1 and Rac2 for NADPH oxidase activation. Exogenous expression of p40(phox) potentiated fMLP-induced O()(2) production and raised the level of O()(2) in unstimulated cells. Collectively, these results provide first direct evidence for reconstituting fMLP-induced O()(2) production in a nonhemopoietic cell line, and demonstrate the requirement of multiple signaling components for optimal activation of NADPH oxidase by a chemoattractant.


Assuntos
N-Formilmetionina Leucil-Fenilalanina/farmacologia , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Neutrófilos/enzimologia , Neutrófilos/imunologia , Fosfoproteínas/genética , Receptores de Formil Peptídeo/fisiologia , Transfecção/métodos , Animais , Células COS , Chlorocebus aethiops , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Humanos , Isoenzimas/genética , Isoenzimas/fisiologia , NADPH Oxidases/antagonistas & inibidores , Neutrófilos/metabolismo , Oxirredução , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/fisiologia , Fosfolipase C beta , Fosfoproteínas/biossíntese , Fosfoproteínas/fisiologia , Proteína Quinase C/genética , Proteína Quinase C/fisiologia , Proteína Quinase C-delta , Receptores de Formil Peptídeo/biossíntese , Receptores de Formil Peptídeo/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Superóxidos/metabolismo , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/fisiologia , Regulação para Cima/genética , Regulação para Cima/imunologia , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/fisiologia , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/fisiologia , Proteína RAC2 de Ligação ao GTP
9.
J Biol Chem ; 277(11): 9268-77, 2002 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11777928

RESUMO

Angiotensin II (Ang II) type 1 receptors (AT1Rs) activate tyrosine kinases, including Src. Whether or not tyrosine kinase activation by AT1R occurs independently of heterotrimeric G protein coupling and, if so, the cellular function of such a mechanism are unknown. To address these questions, we used an AT1aR intracellular second loop mutant, which lacks heterotrimeric G protein coupling (AT1a-i2m). Surprisingly, Ang II-induced Src activation was preserved in AT1a-i2m, which was not attenuated by inhibiting protein kinase C and Ca(2+) or by inhibiting Galpha(i) or Galpha(q) in CHO-K1 cells. By contrast, Ang II-induced Src activation was abolished in a C-terminally truncated AT1a-(1--309), where Ang II-induced inositol phosphate response was preserved. Ang II activates ERKs via a Src-Ras-dependent mechanism in AT1a-i2m. ERKs activated by AT1a-i2m phosphorylate their cytoplasmic targets, including p90(RSK), but fail to translocate into the nucleus or to cause cell proliferation. Ang II-induced nuclear translocation of ERKs by wild type AT1aR was inhibited by overexpression of nuclear exportin Crm-1, while that by AT1a-i2m was restored by leptomycin B, an inhibitor of Crm-1. In summary, while Src and ERKs are activated by Ang II even without heterotrimeric G protein coupling, the carboxyl terminus of the AT1 receptor is required for activation of Src. Interestingly, ERKs activated by heterotrimeric G protein-independent mechanisms fail to phosphorylate nuclear targets due to lack of inhibition of Crm-1-induced nuclear export of ERKs. These results suggest that heterotrimeric G protein-dependent and -independent signaling mechanisms play distinct roles in Ang II-mediated cellular responses.


Assuntos
Transporte Ativo do Núcleo Celular , Proteínas de Ligação a DNA , Proteínas Heterotriméricas de Ligação ao GTP/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores de Angiotensina/fisiologia , Receptores Citoplasmáticos e Nucleares , Fatores de Transcrição , Proteínas ras/fisiologia , Quinases da Família src/fisiologia , Angiotensina II/farmacologia , Animais , Células CHO , Divisão Celular/efeitos dos fármacos , Cricetinae , Ativação Enzimática , Carioferinas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor Tipo 1 de Angiotensina , Transfecção , Proteínas Elk-1 do Domínio ets , Proteína Exportina 1
10.
Mol Pharmacol ; 66(5): 1213-22, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15308762

RESUMO

Formyl peptide receptor-like 1 (FPRL1) is a G protein-coupled receptor that binds natural and synthetic peptides as well as lipoxin A(4) and mediates important biological functions. To facilitate its pharmacological characterization, we screened a compound library and identified a substituted quinazolinone (Quin-C1, 4-butoxy-N-[2-(4-methoxy-phenyl)-4-oxo-1,4-dihydro-2H-quinazolin-3-yl]-benzamide) as a ligand for FPRL1. Quin-C1 induces chemotaxis and secretion of beta-glucuronidase in peripheral blood neutrophils with a potency of approximately 1/1000 of that of the peptide agonist WKYMVm. In studies using transfected rat basophilic leukemia (RBL) cell lines expressing either formyl peptide receptor or FPRL1, Quin-C1 induced enzyme release from RBL-FPRL1 but not RBL-FPR cells. Likewise, Quin-C1 selectively stimulates calcium mobilization in RBL-FPRL1 cells, a response that was markedly inhibited by pertussis toxin. Quin-C1 also stimulates phosphorylation of extracellular signal-regulated protein kinases 1 and 2 and induces internalization of an FPRL1 fused to green fluorescent protein. In degranulation assays, both the FPRL1-selective peptide agonist MMK1 and Quin-C1 exhibited lower efficacy and potency than WKYMVm, with EC(50) values of 7.17 x 10(-8) M and 1.88 x 10(-6) M, respectively, compared with the EC(50) value for WKYMVm (2.29 x 10(-8) M). However, Quin-C1 did not induce neutrophil superoxide generation at up to 100 microM. Based on these results, we conclude that Quin-C1 is a novel nonpeptide ligand that binds to FPRL1 and selectively stimulates FPRL1-mediated functions. Quin-C1 is a prototype of substituted quinazolinones based on which further structural modifications may be made to improve its efficacy and potency for FPRL1.


Assuntos
Benzamidas/farmacologia , Neutrófilos/efeitos dos fármacos , Quinazolinas/farmacologia , Receptores de Formil Peptídeo/agonistas , Animais , Benzamidas/síntese química , Linhagem Celular , Quimiotaxia/efeitos dos fármacos , Interações Medicamentosas , Endocitose/efeitos dos fármacos , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Glucuronidase/metabolismo , Humanos , Ligantes , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neutrófilos/metabolismo , Oligopeptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Quinazolinas/síntese química , Ratos , Receptores de Formil Peptídeo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxidos/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA