Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
Mol Metab ; 10: 87-99, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29453154

RESUMEN

OBJECTIVE: Insulin resistance is a key feature of Type 2 Diabetes (T2D), and improving insulin sensitivity is important for disease management. Allosteric modulation of the insulin receptor (IR) with monoclonal antibodies (mAbs) can enhance insulin sensitivity and restore glycemic control in animal models of T2D. METHODS: A novel human mAb, IRAB-A, was identified by phage screening using competition binding and surface plasmon resonance assays with the IR extracellular domain. Cell based assays demonstrated agonist and sensitizer effects of IRAB-A on IR and Akt phosphorylation, as well as glucose uptake. Lean and diet-induced obese mice were used to characterize single-dose in vivo pharmacological effects of IRAB-A; multiple-dose IRAB-A effects were tested in obese mice. RESULTS: In vitro studies indicate that IRAB-A exhibits sensitizer and agonist properties distinct from insulin on the IR and is translated to downstream signaling and function; IRAB-A bound specifically and allosterically to the IR and stabilized insulin binding. A single dose of IRAB-A given to lean mice rapidly reduced fed blood glucose for approximately 2 weeks, with concomitant reduced insulin levels suggesting improved insulin sensitivity. Phosphorylated IR (pIR) from skeletal muscle and liver were increased by IRAB-A; however, phosphorylated Akt (pAkt) levels were only elevated in skeletal muscle and not liver vs. control; immunochemistry analysis (IHC) confirmed the long-lived persistence of IRAB-A in skeletal muscle and liver. Studies in diet-induced obese (DIO) mice with IRAB-A reduced fed blood glucose and insulinemia yet impaired glucose tolerance and led to protracted insulinemia during a meal challenge. CONCLUSION: Collectively, the data suggest IRAB-A acts allosterically on the insulin receptor acting non-competitively with insulin to both activate the receptor and enhance insulin signaling. While IRAB-A produced a decrease in blood glucose in lean mice, the data in DIO mice indicated an exacerbation of insulin resistance; these data were unexpected and suggested the interplay of complex unknown pharmacology. Taken together, this work suggests that IRAB-A may be an important tool to explore insulin receptor signaling and pharmacology.


Asunto(s)
Sitio Alostérico , Anticuerpos Monoclonales/farmacología , Hipoglucemiantes/farmacología , Receptor de Insulina/agonistas , Células 3T3 , Regulación Alostérica , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Glucemia/metabolismo , Línea Celular Tumoral , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/inmunología , Insulina/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Receptor de Insulina/química , Receptor de Insulina/inmunología , Transducción de Señal
2.
Diabetes ; 66(1): 206-217, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27797911

RESUMEN

A hallmark of type 2 diabetes is impaired insulin receptor (IR) signaling that results in dysregulation of glucose homeostasis. Understanding the molecular origins and progression of diabetes and developing therapeutics depend on experimental models of hyperglycemia, hyperinsulinemia, and insulin resistance. We present a novel monoclonal antibody, IRAB-B, that is a specific, potent IR antagonist that creates rapid and long-lasting insulin resistance. IRAB-B binds to the IR with nanomolar affinity and in the presence of insulin efficiently blocks receptor phosphorylation within minutes and is sustained for at least 3 days in vitro. We further confirm that IRAB-B antagonizes downstream signaling and metabolic function. In mice, a single dose of IRAB-B induces rapid onset of hyperglycemia within 6 h, and severe hyperglycemia persists for 2 weeks. IRAB-B hyperglycemia is normalized in mice treated with exendin-4, suggesting that this model can be effectively treated with a GLP-1 receptor agonist. Finally, a comparison of IRAB-B with the IR antagonist S961 shows distinct antagonism in vitro and in vivo. IRAB-B appears to be a powerful tool to generate both acute and chronic insulin resistance in mammalian models to elucidate diabetic pathogenesis and evaluate therapeutics.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Resistencia a la Insulina/fisiología , Receptor de Insulina/metabolismo , Animales , Western Blotting , Línea Celular , Diabetes Mellitus Tipo 2 , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Humanos , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Péptidos/farmacología , Fosforilación , Unión Proteica , Receptor de Insulina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
3.
J Biol Chem ; 290(41): 24689-704, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26260789

RESUMEN

The efficacy of engaging multiple drug targets using bispecific antibodies (BsAbs) is affected by the relative cell-surface protein levels of the respective targets. In this work, the receptor density values were correlated to the in vitro activity of a BsAb (JNJ-61186372) targeting epidermal growth factor receptor (EGFR) and hepatocyte growth factor receptor (c-MET). Simultaneous binding of the BsAb to both receptors was confirmed in vitro. By using controlled Fab-arm exchange, a set of BsAbs targeting EGFR and c-MET was generated to establish an accurate receptor quantitation of a panel of lung and gastric cancer cell lines expressing heterogeneous levels of EGFR and c-MET. EGFR and c-MET receptor density levels were correlated to the respective gene expression levels as well as to the respective receptor phosphorylation inhibition values. We observed a bias in BsAb binding toward the more highly expressed of the two receptors, EGFR or c-MET, which resulted in the enhanced in vitro potency of JNJ-61186372 against the less highly expressed target. On the basis of these observations, we propose an avidity model of how JNJ-61186372 engages EGFR and c-MET with potentially broad implications for bispecific drug efficacy and design.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Regulación de la Expresión Génica , Terapia Molecular Dirigida , Proteínas Proto-Oncogénicas c-met/inmunología , Proteínas Proto-Oncogénicas c-met/metabolismo , Antígenos de Superficie/química , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Antígenos de Superficie/metabolismo , Línea Celular Tumoral , Receptores ErbB/química , Receptores ErbB/genética , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Modelos Moleculares , Mutación , Fosforilación , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Proteínas Proto-Oncogénicas c-met/química , Proteínas Proto-Oncogénicas c-met/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Methods ; 65(1): 114-26, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23872058

RESUMEN

The Fc variant of IgG2, designated as IgG2σ, was engineered with V234A/G237A /P238S/H268A/V309L/A330S/P331S substitutions to eliminate affinity for Fcγ receptors and C1q complement protein and consequently, immune effector functions. IgG2σ was compared to other previously well-characterized Fc 'muted' variants, including aglycosylated IgG1, IgG2m4 (H268Q/V309L/A330S/P331S, changes to IgG4), and IgG4 ProAlaAla (S228P/L234A/L235A) in its capacity to bind FcγRs and activate various immune-stimulatory responses. In contrast to the previously characterized muted Fc variants, which retain selective FcγR binding and effector functions, IgG2σ shows no detectable binding to the Fcγ receptors in affinity and avidity measurements, nor any detectable antibody-dependent cytotoxicity, phagocytosis, complement activity, or Fc-mediated cytokine release. Moreover, IgG2σ shows minimal immunogenic potential by T-cell epitope analysis. The circulating half-life of IgG2σ in monkeys is extended relative to IgG1 and IgG2, in spite of similar in vitro binding to recombinant FcRn. The three-dimensional structure of the Fc, needed for assessing the basis for the absence of effector function, was compared with that of IgG2 revealing a number of conformational differences near the hinge region of the CH2 domain that result from the amino acid substitutions. Modeling reveals that at least one of the key interactions with FcγRs is disrupted by a conformational change that reorients P329 to a position that prevents it from interacting with conserved W90 and W113 residues of the FcγRs. Inspection of the structure also indicated significant changes to the conformations of D270 and P329 in the CH2 domain that could negatively impact C1q binding. Thus, structural perturbations of the Fc provide a rationale for the loss of function. In toto, these properties of IgG2σ suggest that it is a superior alternative to previously described IgG variants of minimal effector function, for future therapeutic applications of non-immunostimulatory mAb and Fc-fusion platforms.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/química , Factores Inmunológicos/química , Sustitución de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Afinidad de Anticuerpos , Citotoxicidad Celular Dependiente de Anticuerpos , Sitios de Unión , Cristalografía por Rayos X , Citocinas/metabolismo , Células HEK293 , Semivida , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/farmacología , Inmunoglobulina G/genética , Inmunoglobulina G/farmacología , Factores Inmunológicos/genética , Factores Inmunológicos/farmacología , Macaca fascicularis , Masculino , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Secundaria de Proteína , Receptor ErbB-2/inmunología , Receptores de IgG/química
5.
J Mol Recognit ; 25(3): 136-46, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22407977

RESUMEN

In the human adaptation and optimization of a mouse anti-human respiratory syncytial virus neutralizing antibody, affinity assessment was crucial to distinguish among potential candidates and to evaluate whether this correlated with function in vitro and in vivo. This affinity assessment was complicated by the trimeric nature of the antigen target, respiratory syncytial virus F (RSV-F) glycoprotein. In the initial affinity screen, surface plasmon resonance was used to determine the intrinsic binding affinities of anti-RSV-F Fab and immunoglobulin G (IgG) to the extracellular domain of RSV-F. This assessment required minimal biotinylation of the RSV-F protein and design of a capture strategy to minimize avidity effects. Approximately 30 Fabs were selected from three optimization phage display libraries on the basis of an initial ELISA screen. Surface plasmon resonance analysis demonstrated the success of optimization with some candidates from the screened libraries having low picomolar dissociation constants, more than 700-fold tighter than the parental monoclonal antibody (B21M). The affinities of these antibodies were further evaluated by a kinetic exclusion assay, a solution binding technology. One IgG (monoclonal antibody 029) displayed a low picomolar K(D) comparable with that of motavizumab, an RSV antibody in clinical study. Kinetic exclusion assay showed that two other of the matured IgGs (011 and 019) had sub-picomolar dissociation constants that could not be resolved further. We discuss the relevance of these interaction analysis results in the light of recently published data on the mechanism of F-driven viral fusion during paramyxoviral infection and 101F epitope conservation revealed from the recent crystal structure of RSV-F in the post-fusion state.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/química , Anticuerpos Neutralizantes/química , Afinidad de Anticuerpos , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/inmunología , Animales , Biotinilación , Humanos , Concentración de Iones de Hidrógeno , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/inmunología , Fragmentos Fab de Inmunoglobulinas/química , Cinética , Ratones , Biblioteca de Péptidos , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Proteínas Virales de Fusión/química
6.
MAbs ; 2(4): 428-39, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20519961

RESUMEN

We prepared and characterized golimumab (CNTO148), a human IgG1 tumor necrosis factor alpha (TNFα) antagonist monoclonal antibody chosen for clinical development based on its molecular properties. Golimumab was compared with infliximab, adalimumab and etanercept for affinity and in vitro TNFα neutralization. The affinity of golimumab for soluble human TNFα, as determined by surface plasmon resonance, was similar to that of etanercept (18 pM versus 11 pM), greater than that of infliximab (44 pM) and significantly greater than that of adalimumab (127 pM, p=0.018).  The concentration of golimumab necessary to neutralize TNFα-induced E-selectin expression on human endothelial cells by 50% was significantly less than those for infliximab (3.2 fold; p=0.017) and adalimumab (3.3-fold; p=0.008) and comparable to that for etanercept. The conformational stability of golimumab was greater than that of infliximab (primary melting temperature [Tm] 74.8 °C vs. 69.5 °C) as assessed by differential scanning calorimetry.  In addition, golimumab showed minimal aggregation over the intended shelf life when formulated as a high concentration liquid product (100 mg/mL) for subcutaneous administration.  In vivo, golimumab at doses of 1 and 10 mg/kg significantly delayed disease progression in a mouse model of human TNFα-induced arthritis when compared with untreated mice, while infliximab was effective only at 10 mg/kg. Golimumab also significantly reduced histological scores for arthritis severity and cartilage damage, as well as serum levels of pro-inflammatory cytokines and chemokines associated with arthritis. Thus, we have demonstrated that golimumab is a highly stable human monoclonal antibody with high affinity and capacity to neutralize human TNFα in vitro and in vivo.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Artritis/inmunología , Cartílago/efectos de los fármacos , Inmunoglobulina G/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Adalimumab , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales Humanizados/farmacología , Afinidad de Anticuerpos , Artritis/inducido químicamente , Cartílago/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Selectina E/genética , Selectina E/metabolismo , Etanercept , Regulación de la Expresión Génica/efectos de los fármacos , Hibridomas , Inmunoglobulina G/aislamiento & purificación , Mediadores de Inflamación/metabolismo , Infliximab , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Conformación Proteica , Receptores del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/inmunología
7.
Hum Antibodies ; 16(3-4): 117-25, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18334747

RESUMEN

The human CCL2 chemokine is implicated in many chronic inflammatory conditions. In the mouse, there are two CCL2 homologues, CCL2 (MCP-1/JE) and CCL12 (MCP-5). Both are potent monocyte chemoattractants and bind to and activate the same receptor, CCR2. The overlapping activities of these chemokines complicate the design of mouse model studies that are intended to mimic human disease. To study the roles of CCL2 and CCL12, we generated neutralizing antibodies specific to each chemokine. Consistent with binding and affinity analyses, the antibodies specifically inhibited CCL2- or CCL12- mediated Ca(2+) mobilization in THP-1 cells. When tested in nude mice bearing human PANC-1 pancreatic tumor cells in Matrigel plugs, CCL2 and CCL12 antibodies potently inhibited tumor angiogenesis, indicating that both CCL2 and CCL12 may contribute to tumor angiogenesis.


Asunto(s)
Anticuerpos/inmunología , Quimiocina CCL2/inmunología , Proteínas Quimioatrayentes de Monocitos/inmunología , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Línea Celular , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Neovascularización Patológica/prevención & control , Pruebas de Neutralización
8.
Hum Antibodies ; 15(4): 155-62, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17522437

RESUMEN

The generation of anti-variable region monoclonal antibodies (mAbs) against therapeutic antibodies is essential in the pharmacokinetic/pharmacodynamic (PK/PD) assessments of the drugs in clinical study samples. Sandwich EIA and other methods are typically employed to achieve sensitivity and selectivity for the PK/PD analyses. These assays usually require generation of mAb reagents that bind specifically to the therapeutic mAb candidate in non-competing pair combinations. Thus, large panels of anti-variable region mAbs must be generated in an expeditious manner to increase the probability of success. Previously, we described a novel immunization method using type 1 interferons (IFNs) coupled with an agonistic anti-CD40 mAb to drive immune responses (Staquet et al., Human Antibodies 15 (2006), 61-69). This protocol allows for rapid and robust generation of large panels of anti-variable region mAbs. In order to quickly characterize and efficiently identify optimal anti-variable region antibody pairs early in the hybridoma process using crude supernatants, an inexpensive, high-throughput ELISA method was developed. The ability to rapidly identify appropriate mAb pairs will save resources by eliminating the time-consuming and laborious process of subcloning irrelevant hybridomas.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Antígenos CD40/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Región Variable de Inmunoglobulina/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Linfocitos B , Biotecnología/métodos , Femenino , Humanos , Inmunización , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/inmunología , Interferón Tipo I , Ratones , Ratones Endogámicos BALB C , Factores de Tiempo
9.
J Mol Biol ; 349(4): 764-73, 2005 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-15890360

RESUMEN

The cyclin-dependent kinase inhibitors (CKIs) bind to and directly regulate the catalytic activity of cyclin-dependent kinase (Cdk)/cyclin complexes involved in cell cycle control and do not regulate other, closely related Cdks. We showed previously that the CKI, p27, binds to Cdk2/cyclin A though a sequential mechanism that involves folding-on-binding. The first step in the kinetic mechanism is interaction of a small, highly dynamic domain of p27 (domain 1) with the cyclin subunit of the Cdk2/cyclin A complex, followed by much slower binding of a more lengthy and less flexible domain (domain 2) to Cdk2. The second step requires folding of domain 2 into the kinase inhibitory conformation. Rapid binding of p27 domain 1 to cyclin A tethers the inhibitor to the binary Cdk2/cyclin A complex, which reduces the entropic barrier associated with slow binding of domain 2 to the catalytic subunit. We show here that p27/cyclin interactions are an important determinant of p27 specificity towards cell cycle Cdks. We used surface plasmon resonance, limited proteolysis, mass spectrometry, and NMR spectroscopy to study the interaction of p27 with Cdk2/cyclin A, and with another Cdk complex, Cdk5/p25, that is involved in neurodegeneration. Importantly, Cdk5/p35 (the parent complex of Cdk5/p25) is not regulated by p27 in neurons. Our results show that p27 binds to Cdk5 and Cdk2 with similar, slow kinetics. However, p27 fails to interact with p25 within the Cdk5/p25 complex, which we believe prevents formation of a kinetically trapped, inhibited p27/Cdk5/p25 complex in vivo. The helical topology of p25 is very similar to that of cyclin A. However, p25 lacks the MRAIL sequence in one helix that, in the cell cycle cyclins, mediates specific interactions with domain 1 of p21 and p27. Our results strongly suggest that p21 and p27, related Cdk inhibitors, select their cell cycle regulatory Cdk targets by binding specifically to the cyclin subunit of these Cdk/cyclin complexes as a first step in a sequential, folding-on-binding mechanism.


Asunto(s)
Quinasas CDC2-CDC28/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Proteínas de Ciclo Celular/química , División Celular , Ciclina A/química , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina , Quinasa 5 Dependiente de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Humanos , Cinética , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Péptido Hidrolasas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Relación Estructura-Actividad , Especificidad por Sustrato , Resonancia por Plasmón de Superficie , Proteínas Supresoras de Tumor/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA