Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 30(2): 260-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19926832

RESUMEN

OBJECTIVE: The role of myeloid cell cyclooxygenase-2 (COX-2) in the progression of atherosclerosis has not been clearly defined. METHODS AND RESULTS: We investigated the role of COX-2 expressed in the myeloid lineage in the development of atherosclerosis using a myeloid-specific COX-2(-/-) (COX-2(-M/-M)) mouse on a hyperlipidemic apolipoprotein (apo) E(-/-) background (COX-2(-M/-M)/apoE(-/-)). Myeloid COX-2 depletion resulted in significant attenuation of acute inflammation corresponding with decreased PGE(2) levels in an air pouch model. COX-2 depletion in myeloid cells did not influence development of atherosclerosis in COX-2(-M/-M)/apoE(-/-) when compared to apoE(-/-) littermates fed either chow or western diets. The unanticipated lack of contribution of myeloid COX-2 to the development atherosclerosis is not attributable to altered maintenance, differentiation, or mobilization of myeloid and lymphoid populations. Moreover, myeloid COX-2 depletion resulted in unaltered serum prostanoid levels and cellular composition of atherosclerotic lesions of COX-2(-M/-M)/apoE(-/-) mice. CONCLUSIONS: Our results suggest that COX-2 expression in myeloid cells, including macrophages, does not influence the development of atherosclerosis in mice.


Asunto(s)
Enfermedades de la Aorta/enzimología , Apolipoproteínas E/deficiencia , Aterosclerosis/enzimología , Ciclooxigenasa 2/deficiencia , Inflamación/enzimología , Células Mieloides/enzimología , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Células Cultivadas , Ciclooxigenasa 2/genética , Grasas de la Dieta , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/patología , Factor de Necrosis Tumoral alfa/metabolismo
2.
Am J Physiol Gastrointest Liver Physiol ; 298(6): G842-50, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20299600

RESUMEN

Cyclooxygenase-2 (COX-2) is an important regulator of inflammation implicated in the development of a variety of diseases, including inflammatory bowel disease (IBD). However, the regulation of intestinal inflammation by COX-2 is poorly understood. We previously reported that COX-2(-/-) mice fed a cholate-containing high-fat (CCHF) diet had high mortality of unknown mechanisms attributable to severe intestinal inflammation in the ileo-ceco-colic junction that presented characteristics similar to Crohn's disease (CD). To further characterize the role of COX-2 in intestinal inflammation, we established cell-specific conditional COX-2(-/-) mice. Endothelial cell-specific (COX-2(-E/-E)) and myeloid cell-specific (COX-2(-M/-M)) COX-2(-/-) mice, but not wild-type mice, on the CCHF diet developed localized CD-like pathology at the ileo-ceco-colic junction that was associated with cellular infiltration, increased expression of myeloperoxidase and IL-5, and decreased IL-10 expression. The CD-like pathology in COX-2(-E/-E) mice was also accompanied by increased expression of cytokines (IL-6, TNF-alpha, and INF-gamma), compared with wild-type mice and COX-2(-M/-M) mice. In contrast, the ileo-ceco-colic inflammation in COX-2(-M/-M) mice was associated with more pronounced infiltration of granulocytes and macrophages than COX-2(-E/-E) mice. COX-2(-ME/-ME) (COX-2(-M/-M) x COX-2(-E/-E)) mice on the CCHF diet developed CD-like pathology in the ileo-ceco-colic junction reminiscent of total COX-2(-/-) mice on CCHF diet and wild-type mice on CCHF diet treated with COX-2 inhibitor, celecoxib. The pathology of diet-mediated ileo-ceco-colic inflammation in COX-2(-/-) mice offers an excellent model system to elucidate the protective roles of endothelial and myeloid COX-2 and the molecular pathogenesis of CD.


Asunto(s)
Enfermedad de Crohn/metabolismo , Ciclooxigenasa 2/metabolismo , Células Endoteliales/enzimología , Inflamación/enzimología , Células Mieloides/enzimología , Animales , Proteínas de Caenorhabditis elegans , Ciego/patología , Colatos/efectos adversos , Colon Ascendente/patología , Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Ciclooxigenasa 2/genética , Grasas de la Dieta/efectos adversos , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Femenino , Íleon/patología , Inflamación/patología , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Factores de Transcripción
3.
PLoS One ; 9(2): e88684, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24533136

RESUMEN

Previously we reported studies of XMetA, an agonist antibody to the insulin receptor (INSR). We have now utilized phage display to identify XMetS, a novel monoclonal antibody to the INSR. Biophysical studies demonstrated that XMetS bound to the human and mouse INSR with picomolar affinity. Unlike monoclonal antibody XMetA, XMetS alone had little or no agonist effect on the INSR. However, XMetS was a strong positive allosteric modulator of the INSR that increased the binding affinity for insulin nearly 20-fold. XMetS potentiated insulin-stimulated INSR signaling ∼15-fold or greater including; autophosphorylation of the INSR, phosphorylation of Akt, a major enzyme in the metabolic pathway, and phosphorylation of Erk, a major enzyme in the growth pathway. The enhanced signaling effects of XMetS were more pronounced with Akt than with Erk. In cultured cells, XMetS also enhanced insulin-stimulated glucose transport. In contrast to its effects on the INSR, XMetS did not potentiate IGF-1 activation of the IGF-1 receptor. We studied the effect of XMetS treatment in two mouse models of insulin resistance and diabetes. The first was the diet induced obesity mouse, a hyperinsulinemic, insulin resistant animal, and the second was the multi-low dose streptozotocin/high-fat diet mouse, an insulinopenic, insulin resistant animal. In both models, XMetS normalized fasting blood glucose levels and glucose tolerance. In concert with its ability to potentiate insulin action at the INSR, XMetS reduced insulin and C-peptide levels in both mouse models. XMetS improved the response to exogenous insulin without causing hypoglycemia. These data indicate that an allosteric monoclonal antibody can be generated that markedly enhances the binding affinity of insulin to the INSR. These data also suggest that an INSR monoclonal antibody with these characteristics may have the potential to both improve glucose metabolism in insulinopenic type 2 diabetes mellitus and correct compensatory hyperinsulinism in insulin resistant conditions.


Asunto(s)
Anticuerpos Monoclonales/química , Antígenos CD/metabolismo , Glucosa/metabolismo , Receptor de Insulina/metabolismo , Sitio Alostérico , Animales , Péptido C/química , Células CHO , Separación Celular , Cricetinae , Cricetulus , Diabetes Mellitus Tipo 2/metabolismo , Citometría de Flujo , Humanos , Hiperglucemia/metabolismo , Hiperinsulinismo/metabolismo , Insulina/química , Resistencia a la Insulina , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Biblioteca de Péptidos , Fosforilación , Estructura Terciaria de Proteína , Transducción de Señal
4.
MAbs ; 6(1): 262-72, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24423625

RESUMEN

Novel therapies are needed for the treatment of hypoglycemia resulting from both endogenous and exogenous hyperinsulinema. To provide a potential new treatment option, we identified XMetD, an allosteric monoclonal antibody to the insulin receptor (INSR) that was isolated from a human antibody phage display library. To selectively obtain antibodies directed at allosteric sites, panning of the phage display library was conducted using the insulin-INSR complex. Studies indicated that XMetD bound to the INSR with nanomolar affinity. Addition of insulin reduced the affinity of XMetD to the INSR by 3-fold, and XMetD reduced the affinity of the INSR for insulin 3-fold. In addition to inhibiting INSR binding, XMetD also inhibited insulin-induced INSR signaling by 20- to 100-fold. These signaling functions included INSR autophosphorylation, Akt activation and glucose transport. These data indicated that XMetD was an allosteric antagonist of the INSR because, in addition to inhibiting the INSR via modulation of binding affinity, it also inhibited the INSR via modulation of signaling efficacy. Intraperitoneal injection of XMetD at 10 mg/kg twice weekly into normal mice induced insulin resistance. When sustained-release insulin implants were placed into normal mice, they developed fasting hypoglycemia in the range of 50 mg/dl. This hypoglycemia was reversed by XMetD treatment. These studies demonstrate that allosteric monoclonal antibodies, such as XMetD, can antagonize INSR signaling both in vitro and in vivo. They also suggest that this class of allosteric monoclonal antibodies has the potential to treat hyperinsulinemic hypoglycemia resulting from conditions such as insulinoma, congenital hyperinsulinism and insulin overdose.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Hiperinsulinismo Congénito/inmunología , Receptor de Insulina/antagonistas & inhibidores , Anticuerpos de Cadena Única/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/inmunología , Células CHO , Hiperinsulinismo Congénito/tratamiento farmacológico , Hiperinsulinismo Congénito/patología , Cricetinae , Cricetulus , Glucosa/inmunología , Resistencia a la Insulina/inmunología , Ratones , Ratas , Receptor de Insulina/inmunología , Anticuerpos de Cadena Única/farmacología
5.
Diabetes ; 61(5): 1263-71, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22403294

RESUMEN

Many patients with diabetes mellitus (both type 1 and type 2) require therapy to maintain normal fasting glucose levels. To develop a novel treatment for these individuals, we used phage display technology to target the insulin receptor (INSR) complexed with insulin and identified a high affinity, allosteric, human monoclonal antibody, XMetA, which mimicked the glucoregulatory, but not the mitogenic, actions of insulin. Biophysical studies with cultured cells expressing human INSR demonstrated that XMetA acted allosterically and did not compete with insulin for binding to its receptor. XMetA was found to function as a specific partial agonist of INSR, eliciting tyrosine phosphorylation of INSR but not the IGF-IR. Although this antibody activated metabolic signaling, leading to enhanced glucose uptake, it neither activated Erk nor induced proliferation of cancer cells. In an insulin resistant, insulinopenic model of diabetes, XMetA markedly reduced elevated fasting blood glucose and normalized glucose tolerance. After 6 weeks, significant improvements in HbA(1c), dyslipidemia, and other manifestations of diabetes were observed. It is noteworthy that hypoglycemia and weight gain were not observed during these studies. These studies indicate, therefore, that allosteric monoclonal antibodies have the potential to be novel, ultra-long acting, agents for the regulation of hyperglycemia in diabetes.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Glucemia/fisiología , Diabetes Mellitus Experimental/terapia , Receptor de Insulina/agonistas , Animales , Anticuerpos Monoclonales/uso terapéutico , Especificidad de Anticuerpos , Biomarcadores , Células CHO , Células Cultivadas , Cricetinae , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal , Organismos Libres de Patógenos Específicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA