Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Chem Inf Model ; 63(12): 3878-3891, 2023 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-37310029

RESUMEN

Integrins are a family of α/ß heterodimeric cell surface adhesion receptors which are capable of transmitting signals bidirectionally across membranes. They are known for their therapeutic potential in a wide range of diseases. However, the development of integrin-targeting medications has been impacted by unexpected downstream effects including unwanted agonist-like effects. Allosteric modulation of integrins is a promising approach to potentially overcome these limitations. Applying mixed-solvent molecular dynamics (MD) simulations to integrins, the current study uncovers hitherto unknown allosteric sites within the integrin α I domains of LFA-1 (αLß2; CD11a/CD18), VLA-1 (α1ß1; CD49a/CD29), and Mac-1 (αMß2, CD11b/CD18). We show that these pockets are putatively accessible to small-molecule modulators. The findings reported here may provide opportunities for the design of novel allosteric integrin inhibitors lacking the unwanted agonism observed with earlier as well as current integrin-targeting drugs.


Asunto(s)
Antígenos CD18 , Simulación de Dinámica Molecular , Antígenos CD18/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Antígeno de Macrófago-1/metabolismo , Receptores de Superficie Celular
2.
Biochem Pharmacol ; 211: 115504, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36921634

RESUMEN

Integrins are a family of cell surface receptors well-recognized for their therapeutic potential in a wide range of diseases. However, the development of integrin targeting medications has been impacted by unexpected downstream effects, reflecting originally unforeseen interference with the bidirectional signalling and cross-communication of integrins. We here selected one of the most severely affected target integrins, the integrin lymphocyte function-associated antigen-1 (LFA-1, αLß2, CD11a/CD18), as a prototypic integrin to systematically assess and overcome these known shortcomings. We employed a two-tiered ligand-based virtual screening approach to identify a novel class of allosteric small molecule inhibitors targeting this integrin's αI domain. The newly discovered chemical scaffold was derivatized, yielding potent bis-and tris-aryl-bicyclic-succinimides which inhibit LFA-1 in vitro at low nanomolar concentrations. The characterisation of these compounds in comparison to earlier LFA-1 targeting modalities established that the allosteric LFA-1 inhibitors (i) are devoid of partial agonism, (ii) selectively bind LFA-1 versus other integrins, (iii) do not trigger internalization of LFA-1 itself or other integrins and (iv) display oral availability. This profile differentiates the new generation of allosteric LFA-1 inhibitors from previous ligand mimetic-based LFA-1 inhibitors and anti-LFA-1 antibodies, and is projected to support novel immune regulatory regimens selectively targeting the integrin LFA-1. The rigorous computational and experimental assessment schedule described here is designed to be adaptable to the preclinical discovery and development of novel allosterically acting compounds targeting integrins other than LFA-1, providing an exemplary approach for the early characterisation of next generation integrin inhibitors.


Asunto(s)
Antígeno-1 Asociado a Función de Linfocito , Transducción de Señal , Antígeno-1 Asociado a Función de Linfocito/química , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ligandos , Molécula 1 de Adhesión Intercelular/metabolismo
3.
Br J Pharmacol ; 177(12): 2696-2711, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31985813

RESUMEN

BACKGROUND AND PURPOSE: Antibodies targeting cell surface receptors are considered to enable highly selective therapeutic interventions for immune disorders and cancer. Their biological profiles are found, generally, to represent the net effects of antibody-target interactions. The former therapeutic anti-integrin αLß2 antibody efalizumab seems to defeat this paradigm by eliciting, via mechanisms currently unknown, much broader effects than would be predicted based on its target specificity. EXPERIMENTAL APPROACH: To elucidate the mechanisms behind these broad effects, we investigated in primary human lymphocytes in vitro the effects of anti-αLß2 antibodies on the expression of αLß2 as well as unrelated α4 integrins, in comparison to Fab fragments and small-molecule inhibitors. KEY RESULTS: We demonstrate that anti-αLß2 mAbs directly induce the internalization of α4 integrins. The endocytotic phenomenon is a direct consequence of their antibody nature. It is inhibited when monovalent Fab fragments or small-molecule inhibitors are used. It is independent of crosslinking via anti-Fc mAbs and of αLß2 activation. The cross-modulatory effect is unidirectional and not observed in a similar fashion with the α4 integrin antibody natalizumab. CONCLUSION AND IMPLICATIONS: The present study identifies endocytotic cross-modulation as a hitherto unknown non-canonical functionality of anti-αLß2 antibodies. This cross-modulation has the potential to fundamentally alter an antibody's benefit risk profile, as evident with efalizumab. The newly described phenomenon may be of relevance to other therapeutic antibodies targeting cluster-forming receptors. Thus, pharmacologists should be cognizant of this action when investigating such antibodies.


Asunto(s)
Anticuerpos Monoclonales , Antígeno-1 Asociado a Función de Linfocito , Anticuerpos Monoclonales/farmacología , Humanos , Fragmentos Fab de Inmunoglobulinas , Receptores de Superficie Celular
6.
Biochem Pharmacol ; 119: 42-55, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27613223

RESUMEN

The integrin leucocyte function-associated antigen-1 (αLß2, LFA-1) plays crucial roles in T cell adhesion, migration and immunological synapse (IS) formation. Consequently, αLß2 is an important therapeutic target in autoimmunity. Three major classes of αLß2 inhibitors with distinct modes of action have been described to date: Monoclonal antibodies (mAbs), small molecule α/ß I allosteric and small molecule α I allosteric inhibitors. The objective of this study was to systematically compare these three modes of αLß2 inhibition for their αLß2 inhibitory as well as their potential agonist-like effects. All inhibitors assessed were found to potently block αLß2-mediated leucocyte adhesion. None of the inhibitors induced ZAP70 phosphorylation, indicating absence of agonistic outside-in signalling. Paradoxically, however, the α/ß I allosteric inhibitor XVA143 induced conformational changes within αLß2 characteristic for an intermediate affinity state. This effect was not observed with the α I allosteric inhibitor LFA878 or the anti-αLß2 mAb efalizumab. On the other hand, efalizumab triggered the unscheduled internalization of αLß2 in CD4+ and CD8+ T cells while LFA878 and XVA143 did not affect or only mildly reduced αLß2 surface expression, respectively. Moreover, efalizumab, in contrast to the small molecule inhibitors, disturbed the fine-tuned internalization/recycling of engaged TCR/CD3, concomitantly decreasing ZAP70 expression levels. In conclusion, different modes of αLß2 inhibition are associated with fundamentally different biologic effect profiles. The differential established here is expected to provide important translational guidance as novel αLß2 inhibitors will be advanced from bench to bedside.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Integrinas/antagonistas & inhibidores , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales Humanizados , Adhesión Celular , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Integrinas/agonistas , Ratones
7.
Chimia (Aarau) ; 70(12): 864-867, 2016 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-28661358

RESUMEN

Experimental pharmacology is undergoing fundamental changes. This article describes the challenges and opportunities associated with these changes from the perspective of the Swiss Society of Pharmacology (SSEP), the society which aims to advance experimental pharmacology in Switzerland and abroad.


Asunto(s)
Farmacología , Sociedades Científicas/organización & administración , Sociedades Científicas/tendencias , Historia del Siglo XX , Humanos , Farmacología/historia , Farmacología/tendencias , Sociedades Científicas/historia , Suiza
8.
Br J Pharmacol ; 172(20): 4875-87, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26224111

RESUMEN

BACKGROUND AND PURPOSE: The integrin αLß2 plays central roles in leukocyte adhesion and T cell activation, rendering αLß2 an attractive therapeutic target. Compounds with different modes of αLß2 inhibition are in development, currently. Consequently, there is a foreseeable need for bedside assays, which allow assessment of the different effects of diverse types of αLß2 inhibitors in the peripheral blood of treated patients. EXPERIMENTAL APPROACH: Here, we describe a flow cytometry-based technology that simultaneously quantitates αLß2 conformational change upon inhibitor binding, αLß2 expression and T cell activation at the single-cell level in human blood. Two classes of allosteric low MW inhibitors, designated α I and α/ß I allosteric αLß2 inhibitors, were investigated. The first application revealed intriguing inhibitor class-specific profiles. KEY RESULTS: Half-maximal inhibition of T cell activation was associated with 80% epitope loss induced by α I allosteric inhibitors and with 40% epitope gain induced by α/ß I allosteric inhibitors. This differential establishes that inhibitor-induced αLß2 epitope changes do not directly predict the effect on T cell activation. Moreover, we show here for the first time that α/ß I allosteric inhibitors, in contrast to α I allosteric inhibitors, provoked partial downmodulation of αLß2, revealing a novel property of this inhibitor class. CONCLUSIONS AND IMPLICATIONS: The multi-parameter whole blood αLß2 assay described here may enable therapeutic monitoring of αLß2 inhibitors in patients' blood. The assay dissects differential effect profiles of different classes of αLß2 inhibitors.


Asunto(s)
Antígeno-1 Asociado a Función de Linfocito/sangre , Naftalenos/farmacología , Oxazinas/farmacología , Ácidos Ftálicos/farmacología , beta-Alanina/análogos & derivados , Bioensayo , Epítopos/inmunología , Citometría de Flujo , Humanos , Activación de Linfocitos , Linfocitos T/inmunología , beta-Alanina/farmacología
9.
Methods Mol Biol ; 757: 15-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21909903

RESUMEN

Standard adhesion assays measure cell binding either to immobilized ligands or to cell monolayers in flat-well microtiter plates under static conditions. Typically, these test systems require several washing steps to separate adherent from nonadherent cells. Here, we describe an adhesion assay which avoids these washing steps by employing V-bottom 96-well plates. In this assay, fluorescently labeled leukocytes are allowed to adhere to V-well plates coated with soluble ligand for a fixed time. Thereafter, centrifugal force is applied to separate adherent cells from nonadherent cells. Nonadherent cells accumulate in the nadir of the V-shaped wells and are quantified using a fluorometer with a narrow aperture. This simple and reproducible method has been validated with different classes of adhesion molecule families (selectins and integrins) and is adaptable to several other adhesive interactions. The assay format is suitable for screening applications and may also be used for diagnostic testing. The receptor/ligand interaction chosen as an example to describe the assay methodology is the interaction between the integrin lymphocyte function-associated molecule-1 (LFA-1, α(L)ß(2)) and intercellular adhesion molecule-1 (ICAM-1).


Asunto(s)
Adhesión Celular/fisiología , Fluorometría , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Dimetilsulfóxido/farmacología , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Células Jurkat , Leucocitos/metabolismo , Leucocitos Mononucleares/metabolismo , Ligandos , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Unión Proteica , Coloración y Etiquetado
10.
J Biol Chem ; 286(49): 42115-42122, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-21965670

RESUMEN

The activation of α/ß heterodimeric integrins is the result of highly coordinated rearrangements within both subunits. The molecular interactions between the two subunits, however, remain to be characterized. In this study, we use the integrin α(L)ß(2) to investigate the functional role of the C-linker polypeptide that connects the C-terminal end of the inserted (I) domain with the ß-propeller domain on the α subunit and is located at the interface with the ßI domain of the ß chain. We demonstrate that shortening of the C-linker by eight or more amino acids results in constitutively active α(L)ß(2) in which the αI domain is no longer responsive to the regulation by the ßI domain. Despite this intersubunit uncoupling, both I domains remain individually sensitive to intrasubunit conformational changes induced by allosteric modulators. Interestingly, the length and not the sequence of the C-linker appears to be critical for its functionality in α/ß intersubunit communication. Using two monoclonal antibodies (R7.1 and CBR LFA-1/1) we further demonstrate that shortening of the C-linker results in the gradual loss of combinational epitopes that require both the αI and ß-propeller domains for full reactivity. Taken together, our findings highlight the role of the C-linker as a spring-like element that allows relaxation of the αI domain in the resting state and controlled tension of the αI domain during activation, exerted by the ß chain.


Asunto(s)
Integrina alfa1/química , Sitio Alostérico , Animales , Anticuerpos Monoclonales/química , Adhesión Celular , Línea Celular , Membrana Celular/metabolismo , Dimerización , Humanos , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Microscopía Fluorescente/métodos , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Transfección
11.
J Pharmacol Exp Ther ; 330(3): 792-801, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19491325

RESUMEN

There is a pressing need for immunosuppressants with an improved safety profile. The search for novel approaches to blocking T-cell activation led to the development of the selective protein kinase C (PKC) inhibitor AEB071 (sotrastaurin). In cell-free kinase assays AEB071 inhibited PKC, with K(i) values in the subnanomolar to low nanomolar range. Upon T-cell stimulation, AEB071 markedly inhibited in situ PKC catalytic activity and selectively affected both the canonical nuclear factor-kappaB and nuclear factor of activated T cells (but not activator protein-1) transactivation pathways. In primary human and mouse T cells, AEB071 treatment effectively abrogated at low nanomolar concentration markers of early T-cell activation, such as interleukin-2 secretion and CD25 expression. Accordingly, the CD3/CD28 antibody- and alloantigen-induced T-cell proliferation responses were potently inhibited by AEB071 in the absence of nonspecific antiproliferative effects. Unlike former PKC inhibitors, AEB071 did not enhance apoptosis of murine T-cell blasts in a model of activation-induced cell death. Furthermore, AEB071 markedly inhibited lymphocyte function-associated antigen-1-mediated T-cell adhesion at nanomolar concentrations. The mode of action of AEB071 is different from that of calcineurin inhibitors, and AEB071 and cyclosporine A seem to have complementary effects on T-cell signaling pathways.


Asunto(s)
Inmunosupresores/farmacología , Activación de Macrófagos/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/antagonistas & inhibidores , Quinazolinas/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Animales , Antígenos CD28/metabolismo , Calcio/metabolismo , Adhesión Celular/efectos de los fármacos , Citocinas/biosíntesis , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Genes Reporteros , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Células Jurkat , Ratones , Ratones Noqueados , FN-kappa B/efectos de los fármacos , Factores de Transcripción NFATC/metabolismo , Proteína Quinasa C/genética , Receptores de Antígenos de Linfocitos T/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
12.
Mol Pharmacol ; 75(2): 355-62, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18955586

RESUMEN

Natural killer (NK) cells are believed to play an important role in a variety of disease pathologies, including transplant rejection and autoimmunity. None of the therapeutic modalities currently available are known to potently interfere with NK cell activity. Here we demonstrate for the first time that low molecular weight inhibitors of the integrin lymphocyte function-associated antigen-1 (LFA-1) readily block NK cell adhesion, activation, and NK cell-mediated cytolysis in vitro, in contrast to other immunosuppressive agents. These effects were independent of the type of allosteric mechanism by which LFA-1 inhibition was achieved. In addition, we describe a simple, nonradioactive whole-blood assay that should be suitable to monitor NK cell activation in clinical practice. Taken together, our study underlines the importance of LFA-1 in NK cell effector functions and indicates that allosteric LFA-1 inhibitors may become important tools to further elucidate the therapeutic potential of NK cell modulation in immunological diseases.


Asunto(s)
Células Asesinas Naturales/efectos de los fármacos , Antígeno-1 Asociado a Función de Linfocito/fisiología , Fenómenos Biofísicos , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Humanos , Células K562 , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/fisiología , Antígeno-1 Asociado a Función de Linfocito/efectos de los fármacos , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Transducción de Señal/fisiología
13.
J Immunol ; 180(7): 4656-67, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18354189

RESUMEN

Integrin CD18, a component of the LFA-1 complex that also includes CD11a, is essential for Th2, but not Th1, cell homing, but the explanation for this phenomenon remains obscure. In this study, we investigate the mechanism by which Th2 effector responses require the LFA-1 complex. CD11a-deficient T cells showed normal in vitro differentiation and function. However, Th2 cell-dependent allergic lung disease was markedly reduced in CD11a null mice and wild-type mice given LFA-1 inhibitors, whereas control of infection with Leishmania major, a Th1-dependent response, was enhanced. In both disease models, recruitment of IL-4-, but not IFN-gamma-secreting cells to relevant organs was impaired, as was adhesion of Th2 cells in vitro. These diverse findings were explained by the markedly reduced expression of CD29, an alternate homing integrin, on Th2, but not Th1, cells, which precludes Th2 homing in the absence of CD11a. Thus, murine Th1 and Th2 cells use distinct integrins for homing, suggesting novel opportunities for integrin-based therapeutic intervention in diverse human ailments influenced by Th2 cells.


Asunto(s)
Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Regulación de la Expresión Génica/inmunología , Integrinas/metabolismo , Células Th2/citología , Células Th2/inmunología , Alérgenos/inmunología , Animales , Antígeno CD11a/genética , Antígeno CD11a/metabolismo , Células Cultivadas , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Integrina beta1/metabolismo , Leishmania major/inmunología , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/metabolismo , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Ratones Noqueados , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/metabolismo
14.
J Heart Lung Transplant ; 26(7): 724-31, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17613404

RESUMEN

BACKGROUND: Cardiac allograft vasculopathy (CAV) is the pre-eminent cause of late cardiac allograft failure. It is characterized by a concentric intimal hyperplasia, which we designate transplant intimal hyperplasia (TIH). To date, blockade of the adhesion molecule lymphocyte function-associated antigen-1 (LFA-1) has been shown to be effective in preventing TIH in experimental models of transplantation, but only when combined with other immunosuppressants. In this study we explored the impact of monotherapy against LFA-1 in a carotid artery allograft model of TIH. METHODS: B10A(2R) (H-2(h2)) mice were used as donors and C57BL/6 (H-2(b)) mice used as recipients. The recipients were treated with a monoclonal antibody against LFA-1alpha (M17/4) or isotype-matched control immunoglobulin. Grafts were harvested after 35 days and analyzed by histomorphometry and immunohistochemistry. Blood samples were taken and analyzed by differential cell count and alloantibody levels. RESULTS: We found that treatment with M17/4 resulted in a significant reduction in TIH compared with controls. Immunostaining revealed that LFA-1alpha blockade inhibited CD45+ leukocyte infiltration, prevented intimal smooth muscle cell (SMC) proliferation, and preserved the medial SMC population. Finally, we demonstrated a reduction in the serum alloantibody titer in the group treated with anti-LFA-1alpha when compared with controls. CONCLUSIONS: We have demonstrated for the first time that LFA-1alpha blockade on its own can prevent development of TIH in an experimental model. The concept of modulating LFA-1alpha-mediated leukocyte migration and T-cell activation may therefore be of relevance to clinical cardiac transplantation and, as such, represents a potential target for therapeutic intervention against clinical CAV.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Arterias Carótidas/trasplante , Antígeno-1 Asociado a Función de Linfocito/inmunología , Túnica Íntima/patología , Animales , Arterias Carótidas/patología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Hiperplasia/etiología , Hiperplasia/prevención & control , Leucocitos/efectos de los fármacos , Leucocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Trasplante de Tejidos/efectos adversos , Trasplante de Tejidos/patología
15.
Immunology ; 120(3): 315-24, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17140403

RESUMEN

Lymphocyte homing to peripheral lymph nodes is governed by adhesion molecules, including lymphocyte function-associated antigen 1 (LFA-1). Statins are 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors and exert anti-inflammatory effects, e.g. inhibition of LFA-1. It is still not known whether statin compounds are capable of inhibiting lymphocyte homing in vivo. We used a cervical lymph node preparation to study the effects of simvastatin on lymphocyte adhesion to high endothelial venules (HEVs) by means of intravital fluorescence microscopy (IVM). IVM revealed that firm adhesion of lymphocytes to HEV endothelium critically depends on the adhesive function of LFA-1. The number of firmly adherent lymphocytes was reduced by 58% in LFA-1-deficient mice (P < 0.05 versus wild-type controls). As in mutant mice, acute treatment with simvastatin (i.p. injection at 2 hr prior to IVM) inhibited the firm adhesion of lymphocytes to HEV endothelium of wild-type animals by 63% (P < 0.05 versus vehicle-treated wild-type controls). In addition, acute treatment with the synthetic statin-derivate LFA878 also reduced firm lymphocyte adhesion in HEVs by 63% (P > 0.05 versus placebo-treated controls). Histological analysis after a 10-day treatment with simvastatin showed reduced cellularity of cervical lymph nodes, as indicated by a reduction of the relative area of haematoxylin-stained cell nuclei in cervical lymph node cross-sections from 94 +/- 0% in vehicle-treated controls to 77 +/- 3% in simvastatin-treated mice (P < 0.05). We conclude that statin compounds are capable of inhibiting lymphocyte homing to murine peripheral lymph nodes in vivo. This may have novel implications for the treatment of adaptive immune responses, e.g. transplant rejection.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Rodamiento de Leucocito/efectos de los fármacos , Ganglios Linfáticos/inmunología , Linfocitos/efectos de los fármacos , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/inmunología , Recuento de Leucocitos , Vasos Linfáticos/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Naftalenos/farmacología , Cuello , Oxazinas/farmacología , Simvastatina/farmacología
16.
J Leukoc Biol ; 80(2): 215-6, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16735695

RESUMEN

Statins, which inhibit 3-hydroxy-3-methylglutaryl coenzyme-A (HMG-CoA) reductase, are thought to reduce the risk of cancer through the inhibition of Ras farnesylation and serum lipid level. A pleiotropic proinflammatory cytokine, interleukin-18 (IL-18), is reported to exhibit significant antitumor activities through the activation of cytotoxic T lymphocytes and natural killer cells and the inhibition of angiogenesis. Previously, we found that pravastatin, fluvastatin, and simvastatin induced the production of IL-18 in human monocytes. The addition of mevalonate abolished the IL-18 production induced by pravastatin, fluvastatin, and simvastatin, indicating that the IL-18 production might be a result of the inhibition of HMG-CoA reductase. We present a new hypothesis that the production of IL-18 might play roles in the action of statins on cancer.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Interleucina-18/fisiología , Neoplasias/prevención & control , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ácido Mevalónico/farmacología , Modelos Biológicos , Neoplasias/metabolismo
18.
J Leukoc Biol ; 77(3): 400-7, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15618295

RESUMEN

A novel, proinflammatory cytokine, interleukin (IL)-18 production was detected in the medium of human monocytes treated with 3-hydroxy-3-methylglutaryl coenzyme-A (HMG-CoA) reductase inhibitors, pravastatin, and fluvastatin (0.1 and 1 muM) but not with the statin-derived lymphocyte function-associated antigen-1 (LFA-1) inhibitor LFA703, which did not inhibit HMG-CoA reductase. Pravastatin and fluvastatin also induced the production of IL-18, tumor necrosis factor alpha (TNF-alpha) and interferon-gamma (IFN-gamma) in human peripheral blood mononuclear cells (PBMC) in contrast to LFA703. IL-18 production by PBMC is located upstream of the cytokine cascade activated by these statins. The IL-18-induced cytokine production was demonstrated to be dependent on adhesion molecule expression on monocytes. In the absence and presence of lower concentrations (0.1 and 1 ng/ml) of IL-18, pravastatin and fluvastatin inhibited the expression of intercellular adhesion molecule (ICAM)-1 and induced the expression of CD40, whereas LFA703 had no effect. In the presence of higher concentrations (5, 10, and 100 ng/ml) of IL-18, pravastatin, fluvastatin, and LFA703 similarly inhibited the expression of ICAM-1 and CD40 as well as the production of IL-12, TNF-alpha, and IFN-gamma in PBMC. The effects of pravastatin and fluvastatin but not LFA703 were abolished by the addition of mevalonate, indicating the involvement of HMG-CoA reductase in the action of pravastatin and fluvastatin. Thus, the effects of LFA703 were distinct from those of pravastatin and fluvastatin in the presence of lower concentrations of IL-18. It was concluded that LFA703 has the inhibitory effect on an IL-18-initiated immune response without any activation on monocytes.


Asunto(s)
Antígenos CD40/genética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Molécula 1 de Adhesión Intercelular/genética , Interleucina-18/biosíntesis , Monocitos/efectos de los fármacos , Naftalenos/farmacología , Clorometilcetona de Tosilfenilalanila/análogos & derivados , Anticuerpos Monoclonales/farmacología , Antígenos CD40/efectos de los fármacos , Antígenos CD40/metabolismo , Diferenciación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ácidos Grasos Monoinsaturados/farmacología , Fluvastatina , Humanos , Indoles/farmacología , Molécula 1 de Adhesión Intercelular/biosíntesis , Molécula 1 de Adhesión Intercelular/efectos de los fármacos , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Interleucina-18/farmacología , Cinética , Ácido Mevalónico/farmacología , Monocitos/metabolismo , Pravastatina/farmacología , Clorometilcetona de Tosilfenilalanila/farmacología , Factor de Necrosis Tumoral alfa/biosíntesis
19.
Ther Drug Monit ; 26(6): 588-92, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15570181

RESUMEN

Over the past 10-20 years a number of immunosuppressive drugs, such as cyclosporine A, tacrolimus, sirolimus, or mycophenolate mofetil have been approved for clinical use and have been highly successful in preventing or delaying graft rejection. Nevertheless, there is an incessant need for better and safer drugs to improve short-term and long-term outcomes following transplantation. A number of low-molecular-weight molecules that interfere with immune cell functions are in development. These include molecules that inhibit the janus protein tyrosine kinase JAK3, compounds that alter lymphocyte trafficking (the sphingosine-1-phosphate receptor antagonist FTY720), and new malononitrilamides (FK778). All seem to show promising therapeutic potential. Among the biologic agents, there are high expectations for antibodies or recombinant chimeric molecules targeting costimulatory surface molecules or pathways involved in the migration of immune cells. The list of such targets includes the ligand pairs CD28:B7, CD154:CD40, LFA-1:ICAM-1, ICOS:B7RP-1, and VLA-4:VCAM-1. However, the clinical development of drugs for transplantation has proved to be difficult, complex, and time consuming. Therefore, newly emerging drug candidates will also demand better methods for monitoring their efficacy as well as their side effects in vivo. Pharmacokinetics (PK) and pharmacodynamics (PD) are complementary approaches used to select drugs on the basis of their in vivo efficacy as well as safety. Whereas PK monitors the handling of the drug by the body, PD focuses on the biologic effect of the drug on its target. Therefore, PD studies of in vivo efficacy are useful for clinical decisions to determine the optimal dose and type of immunosuppressant. At the preclinical stage, PD is aimed at accelerating the selection of lead compounds via PD-controlled trials in animals. Moreover, PD can help to discover new mechanisms of action for a drug or a drug candidate. However, its full potential has not been used, mainly because of laborious and time-consuming methodology. This review focuses on established and novel PD/PK approaches to assess immunosuppressive compounds in the context of new evolving drugs or drug combinations.


Asunto(s)
Diseño de Fármacos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Tecnología Farmacéutica/métodos , Animales , Humanos , Tecnología Farmacéutica/tendencias
20.
J Biol Chem ; 279(45): 46764-71, 2004 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-15304496

RESUMEN

The integrin lymphocyte function-associated antigen-1 (LFA-1) (alphaLbeta2; CD11a/CD18) plays an important role in leukocyte migration and T cell activation. LFA-1 is inhibited by the cholesterol-lowering drug lovastatin, which binds to an allosteric site of the alphaL I domain termed the lovastatin site (L-site). Here we report for the first time the x-ray structures of the LFA-1 I domain complexed with derivatives of lovastatin optimized for LFA-1 inhibition. This analysis identified two new subpockets within the L-site occupied by chemical groups of the statin derivatives but not by lovastatin itself. Occupancy of these L-site subpockets led to distinct conformational changes in LFA-1, which were detectable by an epitope-monitoring assay. We utilized this assay to demonstrate improved LFA-1 inhibition in human blood in vitro and in blood samples from treated animals ex vivo. Moreover, we demonstrate that the novel lovastatin-derived LFA-1 inhibitor LFA878 exhibits potent anti-inflammatory effects in carrageenan-induced rat paw edema. In summary, the findings reported here extend the understanding of LFA-1 inhibition at the molecular level, allow for the identification and design of LFA-1 inhibitors of further enhanced potency, and support the expectation that LFA-1 inhibitors binding to the L-site will be of therapeutic value in treating inflammatory diseases.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Antígeno-1 Asociado a Función de Linfocito/química , Naftalenos/química , Naftalenos/farmacología , Oxazinas/química , Oxazinas/farmacología , Animales , Sitios de Unión , Sangre/metabolismo , Carragenina/farmacología , Movimiento Celular , Relación Dosis-Respuesta a Droga , Edema , Epítopos/química , Femenino , Humanos , Concentración 50 Inhibidora , Células Jurkat , Leucocitos/citología , Lovastatina/farmacología , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Masculino , Modelos Químicos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Conejos , Ratas , Linfocitos T/citología , Factores de Tiempo , Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...