Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Semin Immunol ; 59: 101604, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35570131

RESUMEN

The complex molecular and cellular biological systems that maintain host homeostasis undergo continuous crosstalk. Complement, a component of innate immunity, is one such system. Initially regarded as a system to protect the host from infection, complement has more recently been shown to have numerous other functions, including involvement in embryonic development, tissue modeling, and repair. Furthermore, the complement system plays a major role in the pathophysiology of many diseases. Through interactions with other plasma cascades, including hemostasis, complement activation leads to the broad host-protective response known as thromboinflammation. Most complement research has been limited to reductionistic models of purified components and cells and their interactions in vitro. However, to study the pathophysiology of complement-driven diseases, including the interaction between the complement system and other inflammatory systems, holistic models demonstrating only minimal interference with complement activity are needed. Here we describe two such models; whole blood anticoagulated with either the thrombin inhibitor lepirudin or the fibrin polymerization peptide blocker GPRP, both of which retain complement activity and preserve the ability of complement to be mutually reactive with other inflammatory systems. For instance, to examine the relative roles of C3 and C5 in complement activation, it is possible to compare the effects of the C3 inhibitor compstatin effects to those of inhibitors of C5 and C5aR1. We also discuss how complement is activated by both pathogen-associated molecular patterns, inducing infectious inflammation caused by organisms such as Gram-negative and Gram-positive bacteria, and by sterile damage-associated molecular patterns, including cholesterol crystals and artificial materials used in clinical medicine. When C3 is inhibited, it is important to determine the mechanism by which inflammation is attenuated, i.e., whether the attenuation derives directly from C3 activation products or via downstream activation of C5, since the mechanism involved may determine the appropriate choice of inhibitor under various conditions. With some exceptions, most inflammatory responses are dependent on C5 and C5aR1; one exception is venous air embolism, in which air bubbles enter the blood circulation and trigger a mainly C3-dependent thromboembolism, with the formation of an active C3 convertase, without a corresponding C5 activation. Under such conditions, an inhibitor of C3 is needed to attenuate the inflammation. Our holistic blood models will be useful for further studies of the inhibition of any complement target, not just C3 or C5. The focus here will be on targeting the critical complement component, activation product, or receptor that is important for the pathophysiology in a variety of disease conditions.


Asunto(s)
Inflamación , Trombosis , Humanos , Proteínas del Sistema Complemento , Activación de Complemento , Complemento C5
2.
Clin Immunol ; 214: 108391, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32229292

RESUMEN

Age-related macular degeneration (AMD) is a leading cause of irreversible vision loss among the elderly population. Genetic studies in susceptible individuals have linked this ocular disease to deregulated complement activity that culminates in increased C3 turnover, retinal inflammation and photoreceptor loss. Therapeutic targeting of C3 has therefore emerged as a promising strategy for broadly intercepting the detrimental proinflammatory consequences of complement activation in the retinal tissue. In this regard, a PEGylated second-generation derivative of the compstatin family of C3-targeted inhibitors is currently in late-stage clinical development as a treatment option for geographic atrophy, an advanced form of AMD which lacks approved therapy. While efficacy has been strongly suggested in phase 2 clinical trials, crucial aspects still remain to be defined with regard to the ocular bioavailability, tissue distribution and residence, and dosing frequency of such inhibitors in AMD patients. Here we report the intraocular distribution and pharmacokinetic profile of the fourth-generation compstatin analog, Cp40-KKK in cynomolgus monkeys following a single intravitreal injection. Using a sensitive surface plasmon resonance (SPR)-based competition assay and ELISA, we have quantified both the amount of inhibitor and the concentration of C3 retained in the vitreous of Cp40-KKK-injected animals. Cp40-KKK displays prolonged intraocular residence, being detected at C3-saturating levels for over 3 months after a single intravitreal injection. Moreover, we have probed the distribution of Cp40-KKK within the ocular tissue by means of immunohistochemistry and highly specific anti-Cp40-KKK antibodies. Both C3 and Cp40-KKK were detected in the retinal tissue of inhibitor-injected animals, with prominent co-localization in the choroid one-month post intravitreal injection. These results attest to the high retinal tissue penetrance and target-driven distribution of Cp40-KKK. Given its subnanomolar binding affinity and prolonged ocular residence, Cp40-KKK constitutes a promising drug candidate for ocular pathologies underpinned by deregulated C3 activation.


Asunto(s)
Complemento C3/antagonistas & inhibidores , Ojo/química , Anciano , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Humanos , Inyecciones Intravítreas , Macaca fascicularis , Retina/química , Factores de Tiempo , Distribución Tisular
3.
Semin Immunol ; 28(3): 208-22, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27321574

RESUMEN

The complement system is a powerful effector arm of innate immunity that typically confers protection from microbial intruders and accumulating debris. In many clinical situations, however, the defensive functions of complement can turn against host cells and induce or exacerbate immune, inflammatory, and degenerative conditions. Although the value of inhibiting complement in a therapeutic context has long been recognized, bringing complement-targeted drugs into clinical use has proved challenging. This important milestone was finally reached a decade ago, yet the clinical availability of complement inhibitors has remained limited. Still, the positive long-term experience with complement drugs and their proven effectiveness in various diseases has reinvigorated interest and confidence in this approach. Indeed, a broad variety of clinical candidates that act at almost any level of the complement activation cascade are currently in clinical development, with several of them being evaluated in phase 2 and phase 3 trials. With antibody-related drugs dominating the panel of clinical candidates, the emergence of novel small-molecule, peptide, protein, and oligonucleotide-based inhibitors offers new options for drug targeting and administration. Whereas all the currently approved and many of the proposed indications for complement-targeted inhibitors belong to the rare disease spectrum, these drugs are increasingly being evaluated for more prevalent conditions. Fortunately, the growing experience from preclinical and clinical use of therapeutic complement inhibitors has enabled a more evidence-based assessment of suitable targets and rewarding indications as well as related technical and safety considerations. This review highlights recent concepts and developments in complement-targeted drug discovery, provides an overview of current and emerging treatment options, and discusses the new milestones ahead on the way to the next generation of clinically available complement therapeutics.


Asunto(s)
Antiinflamatorios/uso terapéutico , Activación de Complemento , Proteínas del Sistema Complemento/inmunología , Enfermedades del Complejo Inmune/terapia , Terapia Molecular Dirigida , Animales , Ensayos Clínicos como Asunto , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Medicina Basada en la Evidencia , Humanos , Enfermedades del Complejo Inmune/inmunología , Inmunidad Innata
4.
Immunobiology ; 221(10): 1046-57, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27353192

RESUMEN

Complement dysregulation is increasingly recognized as an important pathogenic driver in a number of clinical disorders. Complement-triggered pathways intertwine with key inflammatory and tissue destructive processes that can either increase the risk of disease or exacerbate pathology in acute or chronic conditions. The launch of the first complement-targeted drugs in the clinic has undeniably stirred the field of complement therapeutic design, providing new insights into complement's contribution to disease pathogenesis and also helping to leverage a more personalized, comprehensive approach to patient management. In this regard, a rapidly expanding toolbox of complement therapeutics is being developed to address unmet clinical needs in several immune-mediated and inflammatory diseases. Elegant approaches employing both surface-directed and fluid-phase inhibitors have exploited diverse components of the complement cascade as putative points of therapeutic intervention. Targeting C3, the central hub of the system, has proven to be a promising strategy for developing biologics as well as small-molecule inhibitors with clinical potential. Complement modulation at the level of C3 has recently shown promise in preclinical primate models, opening up new avenues for therapeutic intervention in both acute and chronic indications fueled by uncontrolled C3 turnover. This review highlights recent developments in the field of complement therapeutics, focusing on C3-directed inhibitors and alternative pathway (AP) regulator-based approaches. Translational perspectives and considerations are discussed, particularly with regard to the structure-guided drug optimization and clinical advancement of a new generation of C3-targeted peptidic inhibitors.


Asunto(s)
Complemento C3/antagonistas & inhibidores , Complemento C3/inmunología , Inactivadores del Complemento/uso terapéutico , Descubrimiento de Drogas , Inmunoterapia , Producción de Medicamentos sin Interés Comercial , Animales , Estudios Clínicos como Asunto , Activación de Complemento/efectos de los fármacos , Complemento C3/química , Complemento C3/metabolismo , Inactivadores del Complemento/farmacología , Evaluación Preclínica de Medicamentos , Humanos , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Investigación Biomédica Traslacional , Resultado del Tratamiento
5.
Semin Immunol ; 28(3): 285-91, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27021500

RESUMEN

Periodontitis is a dysbiotic inflammatory disease leading to the destruction of the tooth-supporting tissues. Current therapies are not always effective and this prevalent oral disease continues to be a significant health and economic burden. Early clinical studies have associated periodontitis with elevated complement activity. Consistently, subsequent genetic and pharmacological studies in rodents have implicated the central complement component C3 and downstream signaling pathways in periodontal host-microbe interactions that promote dysbiosis and inflammatory bone loss. This review discusses these mechanistic advances and moreover focuses on the compstatin family of C3 inhibitors as a novel approach to treat periodontitis. In this regard, local application of the current lead analog Cp40 was recently shown to block both inducible and naturally occurring periodontitis in non-human primates. These promising results from non-human primate studies and the parallel development of Cp40 for clinical use highlight the feasibility for developing an adjunctive, C3-targeted therapy for human periodontitis.


Asunto(s)
Inactivadores del Complemento/uso terapéutico , Disbiosis/terapia , Boca/inmunología , Periodontitis/terapia , Piridonas/uso terapéutico , Animales , Complemento C3/metabolismo , Complemento C5/metabolismo , Evaluación Preclínica de Medicamentos , Disbiosis/inmunología , Humanos , Boca/microbiología , Periodontitis/inmunología , Primates , Receptor de Anafilatoxina C5a/metabolismo
6.
J Am Soc Mass Spectrom ; 19(1): 55-65, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18293486

RESUMEN

The extracellular fibrinogen-binding protein (Efb), an immunosuppressive and anti-inflammatory protein secreted by Staphylococcus aureus, has been identified as a potent inhibitor of complement-mediated innate immunity. Efb functions by binding to and disrupting the function of complement component 3 (C3). In a recent study, we presented a high-resolution co-crystal structure of the complement inhibitory domain of Efb (Efb-C) bound to its cognate domain (C3d) from human C3 and employed a series of structure/function analyses that provided evidence for an entirely new, conformational change-based mechanism of complement inhibition. To better understand the Efb/C3 complex and its downstream effects on C3 inhibition, we investigated the solvent-accessibility and protein interface of Efb(-C)/C3d using a method of lysine acetylation, proteolytic digestion, and mass spectrometric analysis. Lysine modification in Efb was monitored by the mass increment of lysine-containing fragments. Besides confirming the binding sites observed in co-crystal structure study, the in-solution data presented here suggest additional contacting point(s) between the proteins that were not revealed by crystallography. The results of this study demonstrate that solution-based analysis of protein-protein interactions can provide important complementary information on the nature of protein-protein interactions.


Asunto(s)
Proteínas Bacterianas/química , Complemento C3/química , Lisina/química , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Espectrometría de Masas en Tándem , Acetilación , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión , Datos de Secuencia Molecular , Nanotecnología , Mapeo Peptídico , Proteínas Recombinantes
7.
Nat Biotechnol ; 25(11): 1265-75, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17989689

RESUMEN

The complement system is a central component of innate immunity and bridges the innate to the adaptive immune response. However, it can also turn its destructive capabilities against host cells and is involved in numerous diseases and pathological conditions. Modulation of the complement system has been recognized as a promising strategy in drug discovery, and a large number of therapeutic modalities have been developed. However, successful marketing of complement-targeted drugs has proved to be more difficult than initially expected, and many strategies have been discontinued. The US Food and Drug Administration's approval of the first complement-specific drug, an antibody against complement component C5 (eculizumab; Soliris), in March 2007, was a long-awaited breakthrough in the field. Approval of eculizumab validates the complement system as therapeutic target and might facilitate clinical development of other promising drug candidates.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Inactivadores del Complemento/farmacología , Proteínas del Sistema Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/fisiología , Diseño de Fármacos , Inmunidad Innata/efectos de los fármacos , Anticuerpos Monoclonales/uso terapéutico , Ensayos Clínicos como Asunto , Inactivadores del Complemento/uso terapéutico , Enfermedad/etiología , Evaluación Preclínica de Medicamentos , Humanos
8.
J Med Chem ; 48(1): 274-86, 2005 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-15634022

RESUMEN

We present new findings in our drug discovery effort to develop an anticomplement therapeutic. We have designed several active analogues of compstatin by altering its amino acid composition at positions 4 and 9. The most effective analogues have tryptophan or fused-ring non-natural amino acids at position 4 and alanine or an unbranched single-methyl amino acid at position 9. Twenty-one of these analogues have 2-99-fold higher activities compared to the parent peptide compstatin. The analogue Ac-V4(2Nal)/H9A-NH(2) has the highest inhibitory activity with IC(50) 500 nM. NMR data, through NOE and chemical shift analysis, suggest the presence of interconverting conformers spanning the extended and helical regions of the Ramachandran plot, and they detect a predominant averaged conformer with coil structure and at least one flexible beta-turn, of type I. The fused-ring non-natural amino acids at position 4 contribute to the formation of the hydrophobic cluster of compstatin, which has been previously proposed, together with the beta-turn and a disulfide bridge, to be essential for binding to the target of compstatin, complement component C3. We propose that additional mechanisms may contribute to the structural stability of the analogues and to binding to C3, involving intra- and intermolecular electrostatic interactions of the pi-electron system of side chain aromatic rings. The presence of pi-pi interactions for Trp4-Trp7 was confirmed with a molecular dynamics simulation for the most active analogue with natural amino acids, Ac-V4W/H9A-NH(2). Alanine or aminobutyric acid at position 9 contribute to the weak propensity for helical structure of the residue segment 4-10 of the analogues, which may also play a role in increased activity.


Asunto(s)
Aminoácidos/química , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Sitios de Unión , Células Cultivadas , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Concentración 50 Inhibidora , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Péptidos Cíclicos/metabolismo , Conformación Proteica , Relación Estructura-Actividad
9.
J Immunol ; 169(10): 5962-70, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12421982

RESUMEN

The complement-derived anaphylatoxin, C5a, is a potent phlogistic molecule that mediates its effects by binding to C5a receptor (C5aR; CD88). We now demonstrate specific binding of radiolabeled recombinant mouse C5a to mouse dermal microvascular endothelial cells (MDMEC) with a K(d50) of 3.6 nM and to approximately 15,000-20,000 receptors/cell. Recombinant mC5a competed effectively with binding of [(125)I]rmC5a to MDMEC. Enhanced binding of C5a occurred, as well as increased mRNA for C5aR, after in vitro exposure of MDMEC to LPS, IFN-gamma, or IL-6 in a time- and dose-dependent manner. By confocal microscopy, C5aR could be detected on surfaces of MDMEC using anti-C5aR Ab. In vitro expression of macrophage inflammatory protein-2 (MIP-2) and monocyte chemoattractant protein-1 (MCP-1) by MDMEC was also measured. Exposure of MDMEC to C5a or IL-6 did not result in changes in MIP-2 or MCP-1 production, but initial exposure of MDMEC to IL-6, followed by exposure to C5a, resulted in significantly enhanced production of MIP-2 and MCP-1 (but not TNF-alpha and MIP-1alpha). Although LPS or IFN-gamma alone induced some release of MCP-1 and MIP-2, pre-exposure of these monolayers to LPS or IFN-gamma, followed by addition of C5a, resulted in synergistic production of MIP-2 and MCP-1. Following i.v. infusion of LPS into mice, up-regulation of C5aR occurred in the capillary endothelium of mouse lung, as determined by immunostaining. These results support the hypothesis that C5aR expression on MDMEC and on the microvascular endothelium of lung can be up-regulated, suggesting that C5a in the co-presence of additional agonists may mediate pro-inflammatory effects of endothelial cells.


Asunto(s)
Antígenos CD/biosíntesis , Antígenos CD/fisiología , Complemento C5a/metabolismo , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Receptores de Complemento/biosíntesis , Receptores de Complemento/fisiología , Adyuvantes Inmunológicos/farmacología , Secuencia de Aminoácidos , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Unión Competitiva/inmunología , Células Cultivadas , Quimiocina CCL2/biosíntesis , Quimiocina CXCL2 , Quimiocinas/biosíntesis , Endotelio Vascular/citología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Directa , Regulación de la Expresión Génica/inmunología , Infusiones Intravenosas , Interferón gamma/farmacología , Interleucina-6/farmacología , Radioisótopos de Yodo/metabolismo , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/farmacología , Pulmón/irrigación sanguínea , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Microcirculación/citología , Microcirculación/inmunología , Microcirculación/metabolismo , Microscopía Confocal , Datos de Secuencia Molecular , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/metabolismo , ARN Mensajero/biosíntesis , Receptor de Anafilatoxina C5a , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Regulación hacia Arriba/inmunología , Factor de von Willebrand/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA