Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Am Chem Soc ; 135(1): 22-5, 2013 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-23240907

RESUMEN

In addition to inhibiting the cyclooxygenase (COX)-mediated biosynthesis of prostanoids, various widely used nonsteroidal anti-inflammatory drugs (NSAIDs) enhance endocannabinoid signaling by blocking the anandamide-degrading membrane enzyme fatty acid amide hydrolase (FAAH). The X-ray structure of FAAH in complex with the NSAID carprofen, along with site-directed mutagenesis, enzyme activity assays, and NMR analysis, has revealed the molecular details of this interaction, providing information that may guide the design of dual FAAH-COX inhibitors with superior analgesic efficacy.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/farmacología , Carbazoles/farmacología , Amidohidrolasas/metabolismo , Antiinflamatorios no Esteroideos/química , Sitios de Unión/efectos de los fármacos , Carbazoles/química , Relación Dosis-Respuesta a Droga , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
2.
J Pharm Biomed Anal ; 227: 115256, 2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-36764268

RESUMEN

We recently described C18 fatty acid acylated peptides as a new class of potent long-lasting single-chain RXFP1 agonists that displayed relaxin-like activities in vivo. Early pharmacokinetics and toxicological studies of these stearic acid acylated peptides revealed a relevant oxidative metabolism occurring in dog and minipig, and also seen at a lower extent in monkey and rat. Mass spectrometry combined to NMR spectroscopy studies revealed that the oxidation occurred, unexpectedly, on the stearic acid chain at ω-1, ω-2 and ω-3 positions. Structure-metabolism relationship studies on acylated analogues with different fatty acids lengths (C15-C20) showed that the extent of oxidation was higher with longer chains. The oxidized metabolites could be generated in vitro using liver microsomes and engineered bacterial CYPs. These systems were correlating poorly with in vivo metabolism observed across species; however, the results suggest that this biotransformation pathway might be catalyzed by some unknown CYP enzymes.


Asunto(s)
Sistema Enzimático del Citocromo P-450 , Ácidos Grasos , Animales , Perros , Ratas , Sistema Enzimático del Citocromo P-450/metabolismo , Ácidos Grasos/metabolismo , Redes y Vías Metabólicas , Microsomas Hepáticos/metabolismo , Oxidación-Reducción , Ácidos Esteáricos , Porcinos , Porcinos Enanos/metabolismo , Haplorrinos
3.
Blood ; 116(6): 1002-10, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20466856

RESUMEN

Severe sepsis leads to massive activation of coagulation and complement cascades that could contribute to multiple organ failure and death. To investigate the role of the complement and its crosstalk with the hemostatic system in the pathophysiology and therapeutics of sepsis, we have used a potent inhibitor (compstatin) administered early or late after Escherichia coli challenge in a baboon model of sepsis-induced multiple organ failure. Compstatin infusion inhibited sepsis-induced blood and tissue biomarkers of complement activation, reduced leucopenia and thrombocytopenia, and lowered the accumulation of macrophages and platelets in organs. Compstatin decreased the coagulopathic response by down-regulating tissue factor and PAI-1, diminished global blood coagulation markers (fibrinogen, fibrin-degradation products, APTT), and preserved the endothelial anticoagulant properties. Compstatin treatment also improved cardiac function and the biochemical markers of kidney and liver damage. Histologic analysis of vital organs collected from animals euthanized after 24 hours showed decreased microvascular thrombosis, improved vascular barrier function, and less leukocyte infiltration and cell death, all consistent with attenuated organ injury. We conclude that complement-coagulation interplay contributes to the progression of severe sepsis and blocking the harmful effects of complement activation products, especially during the organ failure stage of severe sepsis is a potentially important therapeutic strategy.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Proteínas Inactivadoras de Complemento/farmacología , Infecciones por Escherichia coli , Insuficiencia Multiorgánica/prevención & control , Péptidos Cíclicos/farmacología , Sepsis , Animales , Biomarcadores/sangre , Coagulación Sanguínea/inmunología , Presión Sanguínea/efectos de los fármacos , Activación de Complemento/efectos de los fármacos , Proteínas Inactivadoras de Complemento/metabolismo , Citocinas/sangre , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/inmunología , Insuficiencia Multiorgánica/sangre , Insuficiencia Multiorgánica/inmunología , Papio , Sepsis/sangre , Sepsis/tratamiento farmacológico , Sepsis/inmunología
4.
Blood ; 116(4): 631-9, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20424189

RESUMEN

Thrombosis is a common complication of end-stage renal disease, particularly in patients on hemodialysis. Although substantial progress has been made in preventing thrombotic complications in various other groups of patients, the mechanisms of thrombosis during hemodialysis require clarification. In this report, we demonstrate that complement activation triggered by hemodialysis biomaterials, and the subsequent generation of the complement anaphylatoxin C5a, results in the expression of functionally active tissue factor (TF) in peripheral blood neutrophils. Because TF is a key initiator of coagulation in vivo, we postulate that the recurring complement activation that occurs during long-term hemodialysis contributes to thrombosis in dialyzed end-stage renal disease patients. Furthermore, we found that complement contributed to the induction of granulocyte colony-stimulating factor, which has been implicated in the pathogenesis of thrombosis in patients treated with the recombinant form of this molecule. Importantly, the inhibition of complement activation attenuated the TF expression and granulocyte colony-stimulating factor induction in blood passing through a hemodialysis circuit, suggesting that the complement system could become a new therapeutic target for preventing thrombosis in patients with chronic renal failure who are maintained on hemodialysis.


Asunto(s)
Complemento C5a/fisiología , Diálisis Renal/efectos adversos , Trombosis/etiología , Anciano , Anafilatoxinas/farmacología , Anafilatoxinas/fisiología , Coagulación Sanguínea/efectos de los fármacos , Células Cultivadas , Activación de Complemento/efectos de los fármacos , Complemento C5a/metabolismo , Complemento C5a/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Fallo Renal Crónico/sangre , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/terapia , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Leucocitos/patología , Masculino , Persona de Mediana Edad , Receptor de Anafilatoxina C5a/metabolismo , Tromboplastina/metabolismo , Factores de Tiempo
5.
Blood ; 116(22): 4395-403, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20625009

RESUMEN

Beyond its role in immunity, complement mediates a wide range of functions in the context of morphogenetic or tissue remodeling processes. Angiogenesis is crucial during tissue remodeling in multiple pathologies; however, the knowledge about the regulation of neovascularization by the complement components is scarce. Here we studied the involvement of complement in pathological angiogenesis. Strikingly, we found that mice deficient in the central complement component C3 displayed increased neovascularization in the model of retinopathy of prematurity (ROP) and in the in vivo Matrigel plug assay. In addition, antibody-mediated blockade of C5, treatment with C5aR antagonist, or C5aR deficiency in mice resulted in enhanced pathological retina angiogenesis. While complement did not directly affect angiogenesis-related endothelial cell functions, we found that macrophages mediated the antiangiogenic activity of complement. In particular, C5a-stimulated macrophages were polarized toward an angiogenesis-inhibitory phenotype, including the up-regulated secretion of the antiangiogenic soluble vascular endothelial growth factor receptor-1. Consistently, macrophage depletion in vivo reversed the increased neovascularization associated with C3- or C5aR deficiency. Taken together, complement and in particular the C5a-C5aR axes are potent inhibitors of angiogenesis.


Asunto(s)
Complemento C3/inmunología , Complemento C5/inmunología , Inmunidad Innata , Neovascularización Patológica/inmunología , Retina/patología , Retinopatía de la Prematuridad/inmunología , Animales , Técnicas de Cultivo de Célula , Línea Celular , Complemento C3/genética , Complemento C5a/inmunología , Eliminación de Gen , Humanos , Recién Nacido , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/patología , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Retina/inmunología , Retinopatía de la Prematuridad/patología , Factores de Crecimiento Endotelial Vascular/inmunología
6.
J Med Chem ; 65(1): 485-496, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34931831

RESUMEN

Inhibitor cystine knot peptides, derived from venom, have evolved to block ion channel function but are often toxic when dosed at pharmacologically relevant levels in vivo. The article describes the design of analogues of ProTx-II that safely display systemic in vivo blocking of Nav1.7, resulting in a latency of response to thermal stimuli in rodents. The new designs achieve a better in vivo profile by improving ion channel selectivity and limiting the ability of the peptides to cause mast cell degranulation. The design rationale, structural modeling, in vitro profiles, and rat tail flick outcomes are disclosed and discussed.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Dolor/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/farmacología , Venenos de Araña/síntesis química , Animales , Degranulación de la Célula/efectos de los fármacos , Cistina/química , Diseño de Fármacos , Calor , Mastocitos/efectos de los fármacos , Modelos Moleculares , Dimensión del Dolor/efectos de los fármacos , Ratas , Venenos de Araña/farmacología
7.
J Med Chem ; 64(4): 2139-2150, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33555858

RESUMEN

The insulin-like peptide human relaxin-2 was identified as a hormone that, among other biological functions, mediates the hemodynamic changes occurring during pregnancy. Recombinant relaxin-2 (serelaxin) has shown beneficial effects in acute heart failure, but its full therapeutic potential has been hampered by its short half-life and the need for intravenous administration limiting its use to intensive care units. In this study, we report the development of long-acting potent single-chain relaxin peptide mimetics. Modifications in the B-chain of relaxin, such as the introduction of specific mutations and the trimming of the sequence to an optimal size, resulted in potent, structurally simplified peptide agonists of the relaxin receptor Relaxin Family Peptide Receptor 1 (RXFP1) (e.g., 54). Introduction of suitable spacers and fatty acids led to the identification of single-chain lipidated peptide agonists of RXFP1, with sub-nanomolar activity, high subcutaneous bioavailability, extended half-lives, and in vivo efficacy (e.g., 64).


Asunto(s)
Lipopéptidos/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores de Péptidos/agonistas , Relaxina/análogos & derivados , Relaxina/farmacología , Secuencia de Aminoácidos , Animales , Enfermedades Cardiovasculares , Línea Celular Tumoral , Células HEK293 , Semivida , Humanos , Lipopéptidos/genética , Lipopéptidos/farmacocinética , Masculino , Simulación de Dinámica Molecular , Estructura Molecular , Mutación , Subunidades de Proteína , Ratas Sprague-Dawley , Relaxina/genética , Relación Estructura-Actividad
8.
J Mol Recognit ; 22(6): 495-505, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19658192

RESUMEN

Compstatin is a 13-residue peptide that inhibits activation of the complement system by binding to the central component C3 and its fragments C3b and C3c. A combination of theoretical and experimental approaches has previously allowed us to develop analogs of the original compstatin peptide with up to 264-fold higher activity; one of these analogs is now in clinical trials for the treatment of age-related macular degeneration (AMD). Here we used functional assays, surface plasmon resonance (SPR), and isothermal titration calorimetry (ITC) to assess the effect of modifications at three key residues (Trp-4, Asp-6, Ala-9) on the affinity and activity of compstatin and its analogs, and we correlated our findings to the recently reported co-crystal structure of compstatin and C3c. The K(D) values for the panel of tested analogs ranged from 10(-6) to 10(-8) M. These differences in binding affinity could be attributed mainly to differences in dissociation rather than association rates, with a >4-fold range in k(on) values (2-10 x 10(5) M(-1) s(-1)) and a k(off) variation of >35-fold (1-37 x 10(-2) s(-1)) being observed. The stability of the C3b-compstatin complex seemed to be highly dependent on hydrophobic effects at position 4, and even small changes at position 6 resulted in a loss of complex formation. Induction of a beta-turn shift by an A9P modification resulted in a more favorable entropy but a loss of binding specificity and stability. The results obtained by the three methods utilized here were highly correlated with regard to the activity/affinity of the analogs. Thus, our analyses have identified essential structural features of compstatin and provided important information to support the development of analogs with improved efficacy.


Asunto(s)
Complemento C3/química , Péptidos Cíclicos/química , Calorimetría/métodos , Indoles/química , Cinética , Modelos Moleculares , Conformación Molecular , Nitrógeno/química , Péptidos/química , Estructura Secundaria de Proteína , Sensibilidad y Especificidad , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie , Triptófano/química , Agua/química
9.
Chem Commun (Camb) ; 53(95): 12814-12817, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-29143042

RESUMEN

N-Acylphosphatidylethanolamine phospholipase D (NAPE-PLD) is a membrane-associated zinc enzyme that catalyzes the hydrolysis of N-acylphosphatidylethanolamines (NAPEs) into fatty acid ethanolamides (FAEs). Here, we describe the identification of the first small-molecule NAPE-PLD inhibitor, the quinazoline sulfonamide derivative 2,4-dioxo-N-[4-(4-pyridyl)phenyl]-1H-quinazoline-6-sulfonamide, ARN19874.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Fosfolipasa D/antagonistas & inhibidores , Quinazolinas/síntesis química , Quinazolinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Sulfonamidas/síntesis química , Sulfonamidas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Fosfolipasa D/metabolismo , Quinazolinas/química , Quinazolinonas , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Sulfonamidas/química
10.
Oncoimmunology ; 6(9): e1326442, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28932632

RESUMEN

Cancer immunotherapy relies upon the ability of T cells to infiltrate tumors. The endothelium constitutes a barrier between the tumor and effector T cells, and the ability to manipulate local vascular permeability could be translated into effective immunotherapy. Here, we show that in the context of adoptive T cell therapy, antitumor T cells, delivered at high enough doses, can overcome the endothelial barrier and infiltrate tumors, a process that requires local production of C3, complement activation on tumor endothelium and release of C5a. C5a, in turn, acts on endothelial cells promoting the upregulation of adhesion molecules and T-cell homing. Genetic deletion of C3 or the C5a receptor 1 (C5aR1), and pharmacological blockade of C5aR1, impaired the ability of T cells to overcome the endothelial barrier, infiltrate tumors, and control tumor progression in vivo, while genetic chimera mice demonstrated that C3 and C5aR1 expression by tumor stroma, and not leukocytes, governs T cell homing, acting on the local endothelium. In vitro, endothelial C3 and C5a expressions were required for endothelial activation by type 1 cytokines. Our data indicate that effective immunotherapy is a consequence of successful homing of T cells in response to local complement activation, which disrupts the tumor endothelial barrier.

11.
J Med Chem ; 49(15): 4616-22, 2006 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-16854067

RESUMEN

Tryptophans at positions 4 and 7 of compstatin, a peptide complement inhibitor, are crucial for its interaction with C3. However, the nature of their involvement has not been studied to date. Here we investigate the molecular forces involved in the C3-compstatin interactions, mediated by aromatic residues, by incorporating in these two positions various tryptophan analogues (5-methyltryptophan, 5-fluorotryptophan, 1-methyltryptophan, and 2-naphthylalanine) and assessing the resulting peptides for activity by enzyme-linked immunosorbent assay (ELISA) and binding by isothermal titration calorimetry (ITC). Of all the compstatin analogues, peptides containing 1-methyltryptophan at position 4 exhibited the highest binding affinity (Kd = 15 nM) and activity (IC50 = 0.205 microM), followed by a peptide containing 5-fluorotryptophan at position 7. Our observations suggest that hydrophobic interactions involving residues at position 4 and the hydrogen bond initiated by the indole nitrogen are primarily responsible and crucial for the increase in activity. These findings have important implications for the design of clinically useful complement inhibitors.


Asunto(s)
Complemento C3/antagonistas & inhibidores , Complemento C3/química , Péptidos Cíclicos/química , Alanina/análogos & derivados , Alanina/síntesis química , Alanina/química , Calorimetría , Ensayo de Inmunoadsorción Enzimática , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Naftalenos/síntesis química , Naftalenos/química , Péptidos Cíclicos/síntesis química , Unión Proteica , Electricidad Estática , Relación Estructura-Actividad , Triptófano/análogos & derivados , Triptófano/síntesis química , Triptófano/química
12.
ACS Chem Biol ; 11(10): 2908-2914, 2016 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-27571266

RESUMEN

The membrane-associated enzyme NAPE-PLD (N-acyl phosphatidylethanolamine specific-phospholipase D) generates the endogenous cannabinoid arachidonylethanolamide and other lipid signaling amides, including oleoylethanolamide and palmitoylethanolamide. These bioactive molecules play important roles in several physiological pathways including stress and pain response, appetite, and lifespan. Recently, we reported the crystal structure of human NAPE-PLD and discovered specific binding sites for the bile acid deoxycholic acid. In this study, we demonstrate that in the presence of this secondary bile acid, the stiffness of the protein measured by elastic neutron scattering increases, and NAPE-PLD is ∼7 times faster to catalyze the hydrolysis of the more unsaturated substrate N-arachidonyl-phosphatidylethanolamine, compared with N-palmitoyl-phosphatidylethanolamine. Chenodeoxycholic acid and glyco- or tauro-dihydroxy conjugates can also bind to NAPE-PLD and drive its activation. The only natural monohydroxy bile acid, lithocholic acid, shows an affinity of ∼20 µM and acts instead as a reversible inhibitor (IC50 ≈ 68 µM). Overall, these findings provide important insights into the allosteric regulation of the enzyme mediated by bile acid cofactors and reveal that NAPE-PLD responds primarily to the number and position of their hydroxyl groups.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Fosfolipasa D/metabolismo , Ácidos y Sales Biliares/química , Cristalografía por Rayos X , Humanos , Cinética , Fosfolipasa D/química , Conformación Proteica , Relación Estructura-Actividad , Especificidad por Sustrato
13.
Exp Neurol ; 274(Pt B): 156-65, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26272754

RESUMEN

Our previous studies revealed that L-type voltage-dependent Ca(2+) channels (Cav1.2 L-VDCCs) are modulated by the neural extracellular matrix backbone, polyanionic glycan hyaluronic acid. Here we used isothermal titration calorimetry and screened a set of peptides derived from the extracellular domains of Cav1.2α1 to identify putative binding sites between the channel and hyaluronic acid or another class of polyanionic glycans, such as heparin/heparan sulfates. None of the tested peptides showed detectable interaction with hyaluronic acid, but two peptides derived from the first pore-forming domain of Cav1.2α1 subunit bound to heparin. At 25 °C the binding of the peptide P7 (MGKMHKTCYN) was at ~50 µM, and that of the peptide P8 (GHGRQCQNGTVCKPGWDGPKHG) was at ~21 µM. The Cav1.2α1 first pore forming segment that contained both peptides maintained a high affinity for heparin (~23 µM), integrating their enthalpic and entropic binding contributions. Interaction between heparin and recombinant as well as native full-length neuronal Cav1.2α1 channels was confirmed using the heparin-agarose pull down assay. Whole cell patch clamp recordings in HEK293 cells transfected with neuronal Cav1.2 channels revealed that enzymatic digestion of highly sulfated heparan sulfates with heparinase 1 affects neither voltage-dependence of channel activation nor the level of steady state inactivation, but did speed up channel inactivation. Treatment of hippocampal cultures with heparinase 1 reduced the firing rate and led to appearance of long-lasting bursts in the same manner as treatment with the inhibitor of L-VDCC diltiazem. Thus, heparan sulfate proteoglycans may bind to and regulate L-VDCC inactivation and network activity.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Heparina/farmacología , Heparitina Sulfato/metabolismo , Neuronas/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Fenómenos Biofísicos/efectos de los fármacos , Fenómenos Biofísicos/genética , Células CHO , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Cricetulus , Diltiazem/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Células HEK293 , Liasa de Heparina/farmacología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Humanos , Microsomas/efectos de los fármacos , Microsomas/metabolismo , Modelos Moleculares , Red Nerviosa/efectos de los fármacos , Neuronas/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Factores de Tiempo , Valina/análogos & derivados , Valina/farmacología
14.
Structure ; 23(3): 598-604, 2015 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-25684574

RESUMEN

The fatty acid ethanolamides (FAEs) are lipid mediators present in all organisms and involved in highly conserved biological functions, such as innate immunity, energy balance, and stress control. They are produced from membrane N-acylphosphatidylethanolamines (NAPEs) and include agonists for G protein-coupled receptors (e.g., cannabinoid receptors) and nuclear receptors (e.g., PPAR-α). Here, we report the crystal structure of human NAPE-hydrolyzing phospholipase D (NAPE-PLD) at 2.65 Å resolution, a membrane enzyme that catalyzes FAE formation in mammals. NAPE-PLD forms homodimers partly separated by an internal ∼ 9-Å-wide channel and uniquely adapted to associate with phospholipids. A hydrophobic cavity provides an entryway for NAPE into the active site, where a binuclear Zn(2+) center orchestrates its hydrolysis. Bile acids bind with high affinity to selective pockets in this cavity, enhancing dimer assembly and enabling catalysis. These elements offer multiple targets for the design of small-molecule NAPE-PLD modulators with potential applications in inflammation and metabolic disorders.


Asunto(s)
Fosfolipasa D/química , Ácidos y Sales Biliares/fisiología , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Enlace de Hidrógeno , Hidrólisis , Modelos Moleculares , Fosfatidiletanolaminas/biosíntesis , Fosfolipasa D/fisiología , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína
15.
Eur J Pharmacol ; 668(1-2): 299-304, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21741965

RESUMEN

Although human mast cells express G protein coupled receptors for the anaphylatoxin C3a, previous studies indicated that C3a causes mast cell degranulation, at least in part, via a C3a receptor-independent mechanism similar to that proposed for polycationic molecules such as compound 48/80. The purpose of the present study was to delineate the receptor specificity of C3a-induced degranulation in human mast cells. We found that C3a, a C3a receptor "superagonist" (E7) and compound 48/80 induced Ca(2+) mobilization and degranulation in a differentiated human mast cell line, LAD2. However, C3a and E7 caused Ca(2+) mobilization in an immature mast cell line, HMC-1 but compound 48/80 did not. We have previously shown that LAD2 cells express MrgX1 and MrgX2 but HMC-1 cells do not. To delineate the receptor specificity for C3a and compound 48/80 further, we generated stable transfectants expressing MrgX1 and MrgX2 in a rodent mast cell line, RBL-2H3 cells. We found that compound 48/80 caused degranulation in RBL-2H3 cells expressing MrgX1 and MrgX2 but C3a did not. By contrast, E7 activated RBL-2H3 cells expressing MrgX2 but not MrgX1. These findings demonstrate that in contrast to previous reports, C3a and compound 48/80 do not use a shared mechanism for mast cell degranulation. It shows that while compound 48/80 utilizes MrgX1 and MrgX2 for mast cell degranulation C3a does not. It further reveals the novel finding that the previously characterized synthetic peptide, C3a receptor "superagonist" E7 activates human mast cells via two mechanisms; one involving the C3a receptor and the other MrgX2.


Asunto(s)
Degranulación de la Célula/efectos de los fármacos , Complemento C3a/metabolismo , Mastocitos/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropéptido/genética , p-Metoxi-N-metilfenetilamina/farmacología , Secuencia de Aminoácidos , Línea Celular , Complemento C3a/agonistas , Complemento C3a/química , Humanos , Mastocitos/citología , Mastocitos/metabolismo , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Proto-Oncogenes Mas , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/agonistas , Receptores de Neuropéptido/metabolismo , Especificidad por Sustrato
16.
Mol Immunol ; 48(4): 481-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21067811

RESUMEN

Compstatin is a 13-residue disulfide-bridged peptide that inhibits a key step in the activation of the human complement system. Compstatin and its derivatives have shown great promise for the treatment of many clinical disorders associated with unbalanced complement activity. To obtain more potent compstatin analogues, we have now performed an N-methylation scan of the peptide backbone and amino acid substitutions at position 13. One analogue (Ac-I[CVW(Me)QDW-Sar-AHRC](NMe)I-NH(2)) displayed a 1000-fold increase in both potency (IC(50) = 62 nM) and binding affinity for C3b (K(D) = 2.3 nM) over that of the original compstatin. Biophysical analysis using surface plasmon resonance and isothermal titration calorimetry suggests that the improved binding originates from more favorable free conformation and stronger hydrophobic interactions. This study provides a series of significantly improved drug leads for therapeutic applications in complement-related diseases, and offers new insights into the structure-activity relationships of compstatin analogues.


Asunto(s)
Proteínas Inactivadoras de Complemento/metabolismo , Péptidos Cíclicos/metabolismo , Secuencia de Aminoácidos , Activación de Complemento/efectos de los fármacos , Proteínas Inactivadoras de Complemento/química , Proteínas Inactivadoras de Complemento/farmacología , Humanos , Cinética , Datos de Secuencia Molecular , Péptidos Cíclicos/química , Unión Proteica/efectos de los fármacos , Termodinámica
17.
Mol Cell Biol ; 30(14): 3596-609, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20457812

RESUMEN

In vitro data have suggested that activation of the inducible T-cell kinase (ITK) requires an interaction with the adaptor protein SLP-76. One means for this interaction involves binding of the ITK SH3 domain to the polyproline-rich (PR) region of SLP-76. However, the biological significance of this association in live cells and the consequences of its disruption have not been demonstrated. Here, we utilized a polyarginine-rich, cell-permeable peptide that represents the portion of the SLP-76 PR region that interacts with the ITK SH3 domain as a competitive inhibitor to disrupt the association between ITK and SLP-76 in live cells. We demonstrate that treatment of cells with this peptide, by either in vitro incubation or intraperitoneal injection of the peptide in mice, inhibits the T-cell receptor (TCR)-induced association between ITK and SLP-76, recruitment and transphosphorylation of ITK, actin polarization at the T-cell contact site, and expression of Th2 cytokines. The inhibition is specific, as indicated by lack of effects by the polyarginine vehicle alone or a scrambled sequence of the cargo peptide. In view of the role of ITK as a regulator of Th2 cytokine expression, the data underscore the significance of ITK as a target for pharmacological intervention.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Citocinas/biosíntesis , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Animales , Unión Competitiva , Humanos , Células Jurkat , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Dominios y Motivos de Interacción de Proteínas , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo
18.
Sci Signal ; 3(109): ra11, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20159852

RESUMEN

Crosstalk between complement and Toll-like receptors (TLRs) coordinates innate immunity. We report a previously unknown immune subversion mechanism involving microbial exploitation of communication between complement and TLRs. Porphyromonas gingivalis, a major oral and systemic pathogen with complement C5 convertase-like activity, synergizes with C5a (fragment of complement protein C5) to increase cyclic adenosine monophosphate (cAMP) concentrations, resulting in suppression of macrophage immune function and enhanced pathogen survival in vitro and in vivo. This synergy required TLR2 signaling, a pertussis toxin- and thapsigargin-sensitive C5a receptor pathway, with protein kinase A and glycogen synthase kinase-3beta as downstream effectors. Antagonistic blockade of the C5a receptor abrogated this evasive strategy and may thus have important therapeutic implications for periodontitis and atherosclerosis, diseases in which P. gingivalis is implicated. This first demonstration of complement-TLR crosstalk for immunosuppressive cAMP signaling indicates that pathogens may not simply undermine complement or TLRs (or both) as separate entities, but may also exploit their crosstalk pathways.


Asunto(s)
Complemento C5/metabolismo , Porphyromonas gingivalis/metabolismo , Receptor Cross-Talk , Receptores Toll-Like/metabolismo , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Macrófagos/inmunología , Ratones , Transducción de Señal
19.
Brain Res ; 1298: 171-7, 2009 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-19410563

RESUMEN

BACKGROUND: Intracerebral hemorrhage (ICH) is associated with neurological injury that may be ameliorated by a neuroprotective strategy targeting the complement cascade. We investigated the role of C5a-receptor antagonist (C5aRA) solely and in combination with C3a-receptor antagonist (C3aRA) following ICH in mice. METHODS: Adult male C57BL/6J mice were randomized to receive vehicle, C5aRA alone or C3aRA and C5aRA 6 and 12 h after ICH, and every 12 h thereafter. A double injection technique was used to infuse 30 microL of autologous whole blood into the right striatum. A final group of mice received a sham procedure consisting only of needle insertion followed by vehicle injections. Brain water content and flow cytometry analysis for leukocyte and microglia infiltration and activation in both hemispheres were measured on day 3 post ICH. Neurological dysfunction was assessed using a Morris water-maze (MWM), a 28-point scale, and a corner test at 6, 12, 24, 48 and 72 h after ICH induction. RESULTS: Neurological deficits were present and comparable in all three cohorts 6 h after ICH. Animals treated with C5aRA and animals treated with combined C3aRA/C5aRA demonstrated significant improvements in neurological function assessed by both the corner turn test and a 28-point neurological scale at 24, 48 and 72 h relative to vehicle-treated animals. Similarly, C5aRA and C3aRA/C5aRA-treated mice demonstrated better spatial memory retention in the Morris water-maze test compared with vehicle-treated animals (C3aRA/C5aRA: 23.4+/-2.0 s p< or =0.0001 versus vehicle: 10.0+/-1.7 s). Relative to vehicle-treated mice, the brain water content in C3aRA/C5aRA-treated mice was significantly decreased in the ipsilateral cortex and ipsilateral striatum (ipsilateral cortex: C3aRA/C5aRA: 0.755403+/-0.008 versus 0.773327+/-0.003 p=0.01 striatum: 0.752273+/-0.007 versus 0.771163+/-0.0036 p=0.02). C5aRA-treated mice and C3aRA/C5aRA-treated mice had a decreased ratio of granulocytes (CD45(+)/CD11b(+)/Ly-6G(+)) in the hemorrhagic versus non-hemorrhagic hemispheres relative to vehicle-treated animals (C5aRA: 1.78+/-0.36 p=0.02 C3aRA/C5aRA: 1.59+/-0.22 p=0.005 versus vehicle: 3.01). CONCLUSIONS: While administration of C5aRA alone provided neuroprotection, combined C3aRA/C5aRA therapy led to synergistic improvements in neurofunctional outcome while reducing inflammatory cell infiltration and brain edema. The results of this study indicate that simultaneous blockade of the C3a and C5a receptors represents a promising neuroprotective strategy in hemorrhagic stroke.


Asunto(s)
Hemorragia Cerebral/tratamiento farmacológico , Citoprotección/efectos de los fármacos , Sinergismo Farmacológico , Granulocitos/efectos de los fármacos , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptores de Complemento/antagonistas & inhibidores , Análisis de Varianza , Animales , Arginina/análogos & derivados , Arginina/farmacología , Conducta Animal/efectos de los fármacos , Compuestos de Bencidrilo/farmacología , Agua Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Química Encefálica/efectos de los fármacos , Hemorragia Cerebral/fisiopatología , Conducta Exploratoria/efectos de los fármacos , Citometría de Flujo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Péptidos Cíclicos/farmacología , Distribución Aleatoria , Conducta Espacial/efectos de los fármacos , Factores de Tiempo
20.
Mol Immunol ; 46(15): 2911-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19665799

RESUMEN

Interactions of gene therapy vectors with human blood components upon intravenous administration have a significant effect on vector efficacy and patient safety. Here we describe methods to evaluate these interactions and their effects in whole human blood, using baculovirus vectors as a model. Opsonisation of baculovirus particles by binding of IgM and C3b was demonstrated, which is likely to be the cause of the significant blood cell-associated virus that was detected. Preventing formation of the complement C5b-9 (membrane attack) complex maintained infectivity of baculovirus particles as shown by studying the effects of two specific complement inhibitors, Compstatin and a C5a receptor antagonist. Formation of macroscopic blood clots after 4h was prevented by both complement inhibitors. Pro- and anti-inflammatory cytokines Il-1beta, IL-6, IL-8 and TNF-alpha were produced at variable levels between volunteers and complement inhibitors showed patient-specific effects on cytokine levels. Whilst both complement inhibitors could play a role in protecting patients from aggressive inflammatory reactions, only Compstatin maintained virus infectivity. We conclude that this ex vivo model, used here for the first time with infectious agents, is a valuable tool in evaluating human innate immune responses to gene therapy vectors or to predict the response of individual patients as part of a clinical trial or treatment. The use of complement inhibitors for therapeutic viruses should be considered on a patient-specific basis.


Asunto(s)
Baculoviridae/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Terapia Genética , Vectores Genéticos/sangre , Vectores Genéticos/inmunología , Inmunidad Innata , Baculoviridae/genética , Complemento C3b/inmunología , Complemento C5a/farmacología , Complejo de Ataque a Membrana del Sistema Complemento/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/inmunología , Vectores Genéticos/genética , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunoensayo , Inmunoglobulina M/sangre , Péptidos Cíclicos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA