Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Neuroinflammation ; 14(1): 88, 2017 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-28427412

RESUMO

BACKGROUND: The majority of Parkinson's disease (PD) cases are sporadic and idiopathic suggesting that this neurodegenerative disorder is the result of both environmental and genetic factors. Stress and neuroinflammation are among the factors being investigated for their possible contributions to PD. Experiments in rodents showed that severe chronic stress can reduce the number of dopaminergic neurons in the substantia nigra pars compacta (SNc); the same cells that are lost in PD. These actions are at least in part mediated by increased oxidative stress. Here, we tested the hypothesis that the interleukin-13 receptor alpha 1 (IL-13Rα1), a cytokine receptor whose activation increases the vulnerability of dopaminergic neurons to oxidative damage, participates in the stress-dependent damage of these neurons. METHODS: Mice were subject to daily sessions of 8 h (acute) stress for 16 weeks (5 days a week), a procedure previously showed to induce loss of dopaminergic neurons in the SNc. The source and the kinetics of interleukin-13 (IL-13), the endogenous ligand of IL-13Rα1, were evaluated 0, 1, 3, 6, and 8 h and at 16 weeks of stress. Identification of IL-13 producing cell-type was performed by immunofluorescent and by in situ hybridization experiments. Markers of oxidative stress, microglia activation, and the number of dopaminergic neurons in IL-13Rα1 knock-out animals (Il13ra1 Y/ - ) and their wild-type littermates (Il13ra1 Y/+ ) were evaluated at 16 weeks of stress and at 20 weeks, following a 4 week non-stressed period and compared to non-stressed mice. RESULTS: IL-13 was expressed in microglial cells within the SN and in a fraction of the tyrosine hydroxylase-positive neurons in the SNc. IL-13 levels were elevated during daily stress and peaked at 6 h. 16 weeks of chronic restraint stress significantly reduced the number of SNc dopaminergic neurons in Il13ra1 Y/+ mice. Neuronal loss at 16 weeks was significantly lower in Il13ra1 Y/- mice. However, the loss of dopaminergic neurons measured at 20 weeks, after 4 weeks of non-stress following the 16 weeks of stress, was similar in Il13ra1 Y/+ and Il13ra1 Y/- mice. CONCLUSIONS: IL-13, a cytokine previously demonstrated to increase the susceptibility of SNc dopaminergic neurons to oxidative stress, is elevated in the SN by restraint stress. Lack of IL-13Rα1 did not prevent nor halted but delayed neuronal loss in the mouse model of chronic restraint stress. IL-13/IL-13Rα1 may represent a target to reduce the rate of DA neuronal loss that can occur during severe chronic restraint stress.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Subunidade alfa1 de Receptor de Interleucina-13/deficiência , Estresse Oxidativo/fisiologia , Estresse Psicológico/metabolismo , Animais , Contagem de Células/métodos , Neurônios Dopaminérgicos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Psicológico/patologia , Substância Negra/metabolismo , Substância Negra/patologia
2.
Mol Pharmacol ; 89(1): 176-86, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26494861

RESUMO

The molecular pharmacology of the G protein-coupled receptors for sphingosine 1-phosphate (S1P) provides important insight into established and new therapeutic targets. A new, potent bitopic S1P3 antagonist, SPM-354, with in vivo activity, has been used, together with S1P3-knockin and S1P3-knockout mice to define the spatial and functional properties of S1P3 in regulating cardiac conduction. We show that S1P3 is a key direct regulator of cardiac rhythm both in vivo and in isolated perfused hearts. 2-Amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol in vivo and S1P in isolated hearts induced a spectrum of cardiac effects, ranging from sinus bradycardia to complete heart block, as measured by a surface electrocardiogram in anesthetized mice and in volume-conducted Langendorff preparations. The agonist effects on complete heart block are absent in S1P3-knockout mice and are reversed in wild-type mice with SPM-354, as characterized and described here. Homologous knockin of S1P3-mCherry is fully functional pharmacologically and is strongly expressed by immunohistochemistry confocal microscopy in Hyperpolarization Activated Cyclic Nucleotide Gated Potassium Channel 4 (HCN4)-positive atrioventricular node and His-Purkinje fibers, with relative less expression in the HCN4-positive sinoatrial node. In Langendorff studies, at constant pressure, SPM-354 restored sinus rhythm in S1P-induced complete heart block and fully reversed S1P-mediated bradycardia. S1P3 distribution and function in the mouse ventricular cardiac conduction system suggest a direct mechanism for heart block risk that should be further studied in humans. A richer understanding of receptor and ligand usage in the pacemaker cells of the cardiac system is likely to be useful in understanding ventricular conduction in health, disease, and pharmacology.


Assuntos
Bloqueio Cardíaco/tratamento farmacológico , Bloqueio Cardíaco/genética , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Animais , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Bloqueio Cardíaco/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Esfingosina-1-Fosfato
3.
Science ; 335(6070): 851-5, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22344443

RESUMO

The lyso-phospholipid sphingosine 1-phosphate modulates lymphocyte trafficking, endothelial development and integrity, heart rate, and vascular tone and maturation by activating G protein-coupled sphingosine 1-phosphate receptors. Here, we present the crystal structure of the sphingosine 1-phosphate receptor 1 fused to T4-lysozyme (S1P(1)-T4L) in complex with an antagonist sphingolipid mimic. Extracellular access to the binding pocket is occluded by the amino terminus and extracellular loops of the receptor. Access is gained by ligands entering laterally between helices I and VII within the transmembrane region of the receptor. This structure, along with mutagenesis, agonist structure-activity relationship data, and modeling, provides a detailed view of the molecular recognition and requirement for hydrophobic volume that activates S1P(1), resulting in the modulation of immune and stromal cell responses.


Assuntos
Receptores de Lisoesfingolipídeo/química , Anilidas/química , Sítios de Ligação , Cristalografia por Raios X , Modelos Moleculares , Muramidase/química , Mutagênese , Organofosfonatos/química , Conformação Proteica , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética
5.
Annu Rev Biochem ; 78: 743-68, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19231986

RESUMO

The sphingosine 1-phosphate (S1P) receptor signaling system is a productive model system. A hydrophobic zwitterionic lysophospholipid ligand with difficult physical properties interacts with five high-affinity G protein-coupled receptors to generate multiple downstream signals. These signals modulate homeostasis and pathology on a steep agonist concentration-response curve. Ligand presence is essential for vascular development and endothelial integrity, while acute increases in ligand concentrations result in cardiac death. Understanding this integrated biochemical system has exemplified the impact of both genetics and chemistry. Developing specific tools with defined biochemical properties for the reversible modulation of signals in real time has been essential to complement insights gained from genetic approaches that may be irreversible and compensated. Despite its knife-edge between life and death, this system, based in part on receptor subtype-selectivity and in part on differential attenuation of deleterious signals, now appears to be on the cusp of meaningful therapy for multiple sclerosis.


Assuntos
Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Animais , Sistema Cardiovascular/embriologia , Sistema Cardiovascular/metabolismo , Humanos , Tecido Linfoide/embriologia , Tecido Linfoide/metabolismo , Esclerose Múltipla/metabolismo , Receptores de Lisoesfingolipídeo/química , Receptores de Lisoesfingolipídeo/genética
6.
Mol Pharmacol ; 74(5): 1308-18, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18708635

RESUMO

Strong evidence exists for interactions of zwitterionic phosphate and amine groups in sphingosine-1 phosphate (S1P) to conserved Arg and Glu residues present at the extracellular face of the third transmembrane domain of S1P receptors. The contribution of Arg(120) and Glu(121) for high-affinity ligand-receptor interactions is essential, because single-point R(120)A or E(121)A S1P(1) mutants neither bind S1P nor transduce S1P function. Because S1P receptors are therapeutically interesting, identifying potent selective agonists with different binding modes and in vivo efficacy is of pharmacological importance. Here we describe a modestly water-soluble highly selective S1P(1) agonist [2-(4-(5-(3,4-diethoxyphenyl)-1,2,4-oxadiazol-3-yl)-2,3-dihydro-1H-inden-1-yl amino) ethanol (CYM-5442)] that does not require Arg(120) or Glu(121) residues for activating S1P(1)-dependent p42/p44 mitogen-activated protein kinase phosphorylation, which defines a new hydrophobic pocket in S1P(1). CYM-5442 is a full agonist in vitro for S1P(1) internalization, phosphorylation, and ubiquitination. It is noteworthy that CYM-5442 was a full agonist for induction and maintenance of S1P(1)-dependent blood lymphopenia, decreasing B lymphocytes by 65% and T lymphocytes by 85% of vehicle. Induction of CYM-5442 lymphopenia was dose- and time-dependent, requiring serum concentrations in the 50 nM range. In vitro measures of S1P(1) activation by CYM-5442 were noncompetitively inhibited by a specific S1P(1) antagonist [(R)-3-amino-(3-hexylphenylamino)-4-oxobutylphosphonic acid (W146)], competitive for S1P, 2-amino-2-(4-octylphenethyl)propane-1,3-diol (FTY720-P), and 5-[4-phenyl-5-(trifluoromethyl)-2-thienyl]-3-[3-(trifluoromethyl)phenyl]-1,2, 4-oxadiazole (SEW2871). In addition, lymphopenia induced by CYM-5442 was reversed by W146 administration or upon pharmacokinetic agonist clearance. Pharmacokinetics in mice also indicated that CYM-5442 partitions significantly in central nervous tissue. These data show that CYM-5442 activates S1P(1)-dependent pathways in vitro and to levels of full efficacy in vivo through a hydrophobic pocket separate from the orthosteric site of S1P binding that is headgroup-dependent.


Assuntos
Indanos/farmacologia , Oxidiazóis/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Animais , Células CHO , Cricetinae , Cricetulus , Ensaio de Imunoadsorção Enzimática , Ligantes , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estrutura Molecular , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Receptores de Lisoesfingolipídeo/metabolismo
7.
Immunol Rev ; 223: 221-35, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18613839

RESUMO

SUMMARY: Modulation of complex functions within the immune system has proven to be surprisingly sensitive to alterations in the lysophospholipid sphingosine 1-phosphate (S1P) receptor-ligand rheostat. This has become increasingly evident from both chemical and genetic manipulation of the S1P system, with pharmacological effects upon lymphoid cells, dendritic cell function, as well as vascular interfaces. The integrated immune system, perhaps as a result of its relatively recent evolutionary ontogeny, has selected for a number of critical control points regulated by five distinct high affinity G-protein-coupled receptor subtypes with a shared ligand, with receptors distributed on lymphocytes, dendritic cells, and endothelium. All of these cellular components of the axis are capable of modulating immune responses in vivo, with the impact on the immune response being very different from classical immunosuppressants, by virtue of selective spatial and temporal sparing of humoral and myeloid elements of host defense. Pharmacological subversion of the S1P rheostat is proving to be clinically efficacious in multiple sclerosis, and both the scope and limitations of therapeutic modulation of the S1P axis in immunotherapy are becoming clearer as understanding of the integrated chemical physiology of the S1P system emerges.


Assuntos
Imunoterapia , Receptores de Lisoesfingolipídeo/imunologia , Transdução de Sinais/imunologia , Animais , Permeabilidade Capilar/imunologia , Movimento Celular/imunologia , Endotélio/imunologia , Endotélio/metabolismo , Humanos , Metabolismo dos Lipídeos/imunologia , Linfócitos/imunologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Esclerose Múltipla/terapia , Receptores de Lisoesfingolipídeo/química , Receptores de Lisoesfingolipídeo/metabolismo , Esfingolipídeos/metabolismo
8.
J Cardiovasc Pharmacol ; 50(6): 660-9, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18091583

RESUMO

Sphingosine-1-phosphate (S1P) has been considered to play an important role in ischemia/reperfusion (I/R) injury. We used SEW2871 (SEW), a novel receptor-selective agonist for S1P1, to elucidate the role of S1P1 in myocardial I/R. Isolated perfused rat hearts exposed to S1P (1 and 10 mM) or SEW (1 and 0.1 mM) were subjected to 30 minutes of global no-flow ischemia and 2 hours of reperfusion. S1P at 1 and 10 mM significantly reduced infarct size and CK release compared with vehicle-control. The effect of 0.1 microM SEW on infarct size was modest. After I/R, S1P at both doses and SEW at 0.1 microM improved developed pressure (LVDP). SEW at 1 mM significantly prolonged the duration of ventricular tachycardia and ventricular fibrillation, leading to irreversible reperfusion tachyarrhythmias in 60% of the hearts. This is the first demonstration of the critical role of the S1P1 receptor in I/R injury.


Assuntos
Arritmias Cardíacas/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Oxidiazóis/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Tiofenos/farmacologia , Compostos de Anilina/farmacologia , Animais , Arritmias Cardíacas/fisiopatologia , Relação Dose-Resposta a Droga , Coração/efeitos dos fármacos , Coração/fisiopatologia , Técnicas In Vitro , Masculino , Modelos Biológicos , Estrutura Molecular , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Oxidiazóis/química , Perfusão , Éteres Fenílicos/farmacologia , Ratos , Ratos Sprague-Dawley , Trocador de Sódio e Cálcio/antagonistas & inibidores , Tiofenos/química , Fatores de Tempo , Função Ventricular Esquerda/efeitos dos fármacos
9.
Trends Immunol ; 28(3): 102-7, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17276731

RESUMO

The lysophospholipid sphingosine 1-phosphate (S1P) is a pleiotropic signaling lipid present constitutively in plasma, and secreted locally at elevated concentrations at sites of inflammation. S1P maintains essential variable homeostatic functions in addition to inducing pathophysiology through the activation of five specific high-affinity G-protein-coupled receptors. Therefore, S1P can function as an extracellular rheostat regulating tonic and acutely evoked functions. Although S1P receptors can regulate lymphoid development and lymphocyte trafficking, and different opinions exist on the roles of receptor agonism and functional antagonism in regulating lymphocyte recirculation, this personal perspective highlights the pivotal control points regulated by constitutive and induced S1P receptor tone at vascular endothelial and lymphatic endothelial barriers, through which S1P agonism impacts on both innate and adaptive immunity. We also emphasize how specific, proof-of-concept chemical tools complement genetic approaches by enabling reversible perturbation of the S1P-S1P(1) receptor axis and, thus, clarifying in vivo mechanisms in the absence of developmental compensations.


Assuntos
Endotélio Vascular/metabolismo , Lisofosfolipídeos/fisiologia , Esfingosina/análogos & derivados , Animais , Antígenos CD/fisiologia , Antígenos de Diferenciação de Linfócitos T/fisiologia , Movimento Celular , Cloridrato de Fingolimode , Humanos , Lectinas Tipo C , Linfócitos/fisiologia , Propilenoglicóis/farmacologia , Receptores de Lisoesfingolipídeo/fisiologia , Transdução de Sinais , Esfingosina/farmacologia , Esfingosina/fisiologia
10.
Nat Chem Biol ; 2(8): 434-41, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16829954

RESUMO

Sphingosine 1-phosphate (S1P, 1) regulates vascular barrier and lymphoid development, as well as lymphocyte egress from lymphoid organs, by activating high-affinity S1P1 receptors. We used reversible chemical probes (i) to gain mechanistic insights into S1P systems organization not accessible through genetic manipulations and (ii) to investigate their potential for therapeutic modulation. Vascular (but not airway) administration of the preferred R enantiomer of an in vivo-active chiral S1P1 receptor antagonist induced loss of capillary integrity in mouse skin and lung. In contrast, the antagonist did not affect the number of constitutive blood lymphocytes. Instead, alteration of lymphocyte trafficking and phenotype required supraphysiological elevation of S1P1 tone and was reversed by the antagonist. In vivo two-photon imaging of lymph nodes confirmed requirements for obligate agonism, and the data were consistent with the presence of a stromal barrier mechanism for gating lymphocyte egress. Thus, chemical modulation reveals differences in S1P-S1P1 'set points' among tissues and highlights both mechanistic advantages (lymphocyte sequestration) and risks (pulmonary edema) of therapeutic intervention.


Assuntos
Anilidas/farmacologia , Linfócitos/efeitos dos fármacos , Organofosfonatos/farmacologia , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Anilidas/administração & dosagem , Anilidas/síntese química , Animais , Células CHO , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Células Cultivadas , Cricetinae , Modelos Animais de Doenças , Azul Evans/química , Humanos , Linfonodos/efeitos dos fármacos , Linfócitos/metabolismo , Lisofosfolipídeos/química , Lisofosfolipídeos/farmacologia , Lisofosfolipídeos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Organofosfonatos/administração & dosagem , Organofosfonatos/síntese química , Fenótipo , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/diagnóstico , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Esfingosina/química , Esfingosina/farmacologia , Esfingosina/fisiologia , Estereoisomerismo
11.
Nat Immunol ; 6(12): 1228-35, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16273098

RESUMO

Sphingosine 1-phosphate type 1 (S1P(1)) receptor agonists cause sequestration of lymphocytes in secondary lymphoid organs by a mechanism that is not well understood. One hypothesis proposes that agonists act as 'functional antagonists' by binding and internalizing S1P(1) receptors on lymphocytes; a second hypothesis proposes instead that S1P(1) agonists act on endothelial cells to prevent lymphocyte egress from lymph nodes. Here, two-photon imaging of living T cells in explanted lymph nodes after treatment with S1P(1) agonists or antagonists has provided insight into the mechanism by which S1P(1) agonists function. The selective S1P(1) agonist SEW2871 caused reversible slowing and 'log-jamming' of T cells between filled medullary cords and empty sinuses, whereas motility was unaltered in diffuse cortex. Removal or antagonist competition of SEW2871 permitted recovery of T cell motility in the parenchyma of the medulla and resumption of migration across the stromal endothelial barrier, leading to refilling of sinuses. Our results provide visualization of transendothelial migration of T cells into lymphatic sinuses and suggest that S1P(1) agonists act mainly on endothelial cell S1P(1) receptors to inhibit lymphocyte migration.


Assuntos
Inibição de Migração Celular , Movimento Celular/fisiologia , Linfonodos/fisiologia , Vasos Linfáticos/fisiologia , Lisofosfolipídeos/metabolismo , Oxidiazóis/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Esfingosina/análogos & derivados , Subpopulações de Linfócitos T/fisiologia , Tiofenos/farmacologia , Animais , Células Cultivadas , Linfonodos/citologia , Vasos Linfáticos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Esfingosina/metabolismo , Subpopulações de Linfócitos T/efeitos dos fármacos
12.
Proc Natl Acad Sci U S A ; 102(26): 9270-5, 2005 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-15968000

RESUMO

Pulmonary pathologies including adult respiratory distress syndrome are characterized by disruption of pulmonary integrity and edema compromising respiratory function. Sphingosine 1-phosphate (S1P) is a lipid mediator synthesized and/or stored in mast cells, platelets, and epithelial cells, with production up-regulated by the proinflammatory cytokines IL-1 and TNF. S1P administration via the airways but not via the vasculature induces lung leakage. Using receptor-null mice, we show that S1P, acting on S1P3 receptor expressed on both type I and type II alveolar epithelial cells but not vascular endothelium, induces pulmonary edema by acute tight junction opening. WT but not S1P3-null mice showed disruption of pulmonary epithelial tight junctions and the appearance of paracellular gaps between epithelial cells by electron microscopy within 1 h of airways exposure to S1P. We further show by fluorescence microscopy that S1P induced rapid loss of ZO-1 reactivity, an essential component of the cytoplasmic plaque associated with tight junctions, as well as of the tetraspannin Claudin-18, an integral membrane organizer of tight junctions. S1P shows synergistic activity with the proinflammatory cytokine TNF, showing both pulmonary edema and mortality at subthreshold S1P doses. Specifically, preexposure of mice to subthreshold doses of TNF, which alone induced no lung edema, exacerbated S1P-induced edema and impaired survival. S1P, acting through S1P3, regulates epithelial integrity and acts additively with TNF in compromising respiratory barrier function. Because S1P3-null mice are resistant to S1P-induced pulmonary leakage, either alone or in the presence of TNF, S1P3 antagonism may be useful in protecting epithelial integrity in pulmonary disease.


Assuntos
Células Epiteliais/metabolismo , Pulmão/metabolismo , Receptores de Lisoesfingolipídeo/fisiologia , Junções Íntimas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Cromatografia Líquida , Claudinas , Citoplasma/metabolismo , Endotélio Vascular/metabolismo , Epitélio/metabolismo , Guanosina Trifosfato/metabolismo , Homozigoto , Inflamação , Interleucina-1/metabolismo , Ligantes , Pneumopatias/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica , Microscopia de Fluorescência , Modelos Biológicos , Permeabilidade , Ligação Proteica , RNA Mensageiro/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Fatores de Tempo
13.
Chem Biol ; 12(6): 703-15, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15975516

RESUMO

The essential role of the sphingosine 1-phosphate (S1P) receptor S1P(1) in regulating lymphocyte trafficking was demonstrated with the S1P(1)-selective nanomolar agonist, SEW2871. Despite its lack of charged headgroup, the tetraaromatic compound SEW2871 binds and activates S1P(1) through a combination of hydrophobic and ion-dipole interactions. Both S1P and SEW2871 activated ERK, Akt, and Rac signaling pathways and induced S1P(1) internalization and recycling, unlike FTY720-phosphate, which induces receptor degradation. Agonism with receptor recycling is sufficient for alteration of lymphocyte trafficking by S1P and SEW2871. S1P(1) modeling and mutagenesis studies revealed that residues binding the S1P headgroup are required for kinase activation by both S1P and SEW2871. Therefore, SEW2871 recapitulates the action of S1P in all the signaling pathways examined and overlaps in interactions with key headgroup binding receptor residues, presumably replacing salt-bridge interactions with ion-dipole interactions.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Sondas Moleculares/metabolismo , Oxidiazóis/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tiofenos/farmacologia , Animais , Sítios de Ligação , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetinae , Ativação Enzimática/efeitos dos fármacos , Humanos , Ligantes , Lisofosfolipídeos/química , Lisofosfolipídeos/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Modelos Moleculares , Sondas Moleculares/química , Sondas Moleculares/farmacologia , Mutação/genética , Oxidiazóis/química , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptores de Lisoesfingolipídeo/química , Receptores de Lisoesfingolipídeo/genética , Esfingosina/análogos & derivados , Esfingosina/química , Esfingosina/farmacologia , Tiofenos/química , Proteínas rac de Ligação ao GTP/metabolismo
14.
J Biol Chem ; 279(14): 13839-48, 2004 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-14732717

RESUMO

Sphingosine 1-phosphate (S1P) influences heart rate, coronary artery caliber, endothelial integrity, and lymphocyte recirculation through five related high affinity G-protein-coupled receptors. Inhibition of lymphocyte recirculation by non-selective S1P receptor agonists produces clinical immunosuppression preventing transplant rejection but is associated with transient bradycardia. Understanding the contribution of individual receptors has been limited by the embryonic lethality of the S1P(1) knock-out and the unavailability of selective agonists or antagonists. A potent, S1P(1)-receptor selective agonist structurally unrelated to S1P was found to activate multiple signals triggered by S1P, including guanosine 5'-3-O-(thio)triphosphate binding, calcium flux, Akt and ERK1/2 phosphorylation, and stimulation of migration of S1P(1)- but not S1P(3)-expressing cells in vitro. The agonist also alters lymphocyte trafficking in vivo. Use of selective agonism together with deletant mice lacking S1P(3) receptor reveals that agonism of S1P(1) receptor alone is sufficient to control lymphocyte recirculation. Moreover, S1P(1) receptor agonist plasma levels are causally associated with induction and maintenance of lymphopenia. S1P(3), and not S1P(1), is directly implicated in sinus bradycardia. The sustained bradycardia induced by S1P receptor non-selective immunosuppressive agonists in wild-type mice is abolished in S1P(3)-/- mice, whereas S1P(1)-selective agonist does not produce bradycardia. Separation of receptor subtype usage for control of lymphocyte recirculation and heart rate may allow the identification of selective immunosuppressive S1P(1) receptor agonists with an enhanced therapeutic window. S1P(1)-selective agonists will be of broad utility in understanding cell functions in vitro, and vascular physiology in vivo, and the success of the chemical approach for S1P(1) suggests that selective tools for the resolution of function across this broad lipid receptor family are now possible.


Assuntos
Movimento Celular/imunologia , Frequência Cardíaca/fisiologia , Linfócitos/citologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Células CHO , Cálcio/metabolismo , Movimento Celular/efeitos dos fármacos , Cricetinae , Cloridrato de Fingolimode , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Imunossupressores/farmacologia , Sistema Linfático/citologia , Linfopenia/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fosforilação/efeitos dos fármacos , Propilenoglicóis/farmacologia , Proteínas Quinases/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores de Lisofosfolipídeos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Esfingosina/análogos & derivados
15.
J Biol Chem ; 277(34): 30454-62, 2002 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-12048196

RESUMO

We have previously described a new aspect of the Inhibitor of Apoptosis (IAP) family of proteins anti-apoptotic activity that involves the TAK1/JNK1 signal transduction pathway (1,2). Our findings suggest the existence of a novel mechanism that regulates the anti-apoptotic activity of IAPs that is separate from caspase inhibition but instead involves TAK1-mediated activation of JNK1. In a search for proteins involved in the XIAP/TAK1/JNK1 signaling pathway we isolated by yeast two-hybrid screening a novel X chromosome-linked IAP (XIAP)-interacting protein that we called ILPIP (hILP-Interacting Protein). Whereas ILPIP moderately activates JNK family members when expressed alone, it strongly enhances XIAP-mediated activation of JNK1, JNK2, and JNK3. The expression of a catalytically inactive mutant of TAK1 blocked XIAP/ILPIP synergistic activation of JNK1 thereby implicating TAK1 in this signaling pathway. ILPIP moderately protects against interleukin-1beta converting enzyme- or Fas-induced apoptosis and significantly potentiates the anti-apoptotic activity of XIAP. In vivo co-precipitation experiments show that both ILPIP and XIAP interact with TAK1 and tumor necrosis factor receptor-associated factor 6. Finally, expression of ILPIP did not affect the ability of XIAP to inhibit caspase activation, further supporting the idea that XIAP protection against apoptosis is achieved by two separate mechanisms: one requiring JNK1 activation and a second involving caspase inhibition.


Assuntos
Apoptose , MAP Quinase Quinase Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Caspases/metabolismo , Células Cultivadas , Ativação Enzimática , Humanos , Proteína Quinase 8 Ativada por Mitógeno , Dados de Sequência Molecular , Proteínas Quinases/fisiologia , Transdução de Sinais , Fator 6 Associado a Receptor de TNF , Técnicas do Sistema de Duplo-Híbrido , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X
16.
Mol Cell Biol ; 22(6): 1754-66, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11865055

RESUMO

The antiapoptotic properties of the inhibitor of apoptosis (IAP) family of proteins have been linked to caspase inhibition. We have previously described an alternative mechanism of XIAP inhibition of apoptosis that depends on the selective activation of JNK1. Here we report that two other members of the IAP family, NAIP and ML-IAP, both activate JNK1. Expression of catalytically inactive JNK1 blocks NAIP and ML-IAP protection against ICE- and TNF-alpha-induced apoptosis, indicating that JNK1 activation is necessary for the antiapoptotic effect of these proteins. The MAP3 kinase, TAK1, appears to be an essential component of this antiapoptotic pathway since IAP-mediated activation of JNK1, as well as protection against TNF-alpha- and ICE-induced apoptosis, is inhibited when catalytically inactive TAK1 is expressed. In addition, XIAP, NAIP, and JNK1 bind to TAK1. Importantly, expression of catalytically inactive TAK1 did not affect XIAP inhibition of caspase activity. These data suggest that XIAP's antiapoptotic activity is achieved by two separate mechanisms: one requiring TAK1-dependent JNK1 activation and the second involving caspase inhibition.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose/fisiologia , Inibidores de Caspase , Proteínas de Insetos/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , MAP Quinase Quinase 4 , MAP Quinase Quinase Quinases/metabolismo , Proteínas Associadas aos Microtúbulos , Proteínas Proto-Oncogênicas c-bcl-2 , Transdução de Sinais/fisiologia , Apoptose/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Caspase 1/farmacologia , Caspases/metabolismo , Linhagem Celular , Proteínas Cromossômicas não Histona/metabolismo , Ativação Enzimática/efeitos dos fármacos , Proteína gp120 do Envelope de HIV/metabolismo , Humanos , Proteínas Inibidoras de Apoptose , Proteínas de Insetos/farmacologia , Rim/citologia , Rim/metabolismo , MAP Quinase Quinase 7 , Proteína Quinase 8 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteína Inibidora de Apoptose Neuronal , Ligação Proteica , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Survivina , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X , Proteína X Associada a bcl-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA