Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Chem ; 60(5): 747-57, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24486646

RESUMEN

BACKGROUND: Acute kidney injury (AKI) affects 45% of critically ill patients, resulting in increased morbidity and mortality. The diagnostic standard, plasma creatinine, is nonspecific and may not increase until days after injury. There is significant need for a renal-specific AKI biomarker detectable early enough that there would be a potential window for therapeutic intervention. In this study, we sought to identify a renal-specific biomarker of AKI. METHODS: We analyzed gene expression data from normal mouse tissues to identify kidney-specific genes, one of which was Miox. We generated monoclonal antibodies to recombinant myo-inositol oxygenase (MIOX) and developed an immunoassay to quantify MIOX in plasma. The immunoassay was tested in animals and retrospectively in patients with and without AKI. RESULTS: Kidney tissue specificity of MIOX was supported by Western blot. Immunohistochemistry localized MIOX to the proximal renal tubule. Serum MIOX, undetectable at baseline, increased 24 h following AKI in mice. Plasma MIOX was increased in critically ill patients with AKI [mean (SD) 12.4 (4.3) ng/mL, n = 42] compared with patients without AKI [0.5 (0.3) ng/mL, n = 17] and was highest in patients with oliguric AKI [20.2 (7.5) ng/mL, n = 23]. Plasma MIOX increased 54.3 (3.8) h before the increase in creatinine. CONCLUSIONS: MIOX is a renal-specific, proximal tubule protein that is increased in serum of animals and plasma of critically ill patients with AKI. MIOX preceded the increases in creatinine concentration by approximately 2 days in human patients. Large-scale studies are warranted to further investigate MIOX as an AKI biomarker.


Asunto(s)
Lesión Renal Aguda/sangre , Inmunoensayo/métodos , Oxigenasas/sangre , Lesión Renal Aguda/enzimología , Animales , Anticuerpos Monoclonales/inmunología , Biomarcadores/sangre , Western Blotting , Enfermedad Crítica , Femenino , Humanos , Inmunohistoquímica , Inositol-Oxigenasa , Masculino , Ratones , Oxigenasas/genética , Oxigenasas/inmunología , ARN Mensajero/genética
2.
Am J Physiol Gastrointest Liver Physiol ; 306(4): G301-9, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24356886

RESUMEN

Xenin-25 (Xen) is a neurotensin-related peptide secreted by a subset of glucose-dependent insulinotropic polypeptide (GIP)-producing enteroendocrine cells. In animals, Xen regulates gastrointestinal function and glucose homeostasis, typically by initiating neural relays. However, little is known about Xen action in humans. This study determines whether exogenously administered Xen modulates gastric emptying and/or insulin secretion rates (ISRs) following meal ingestion. Fasted subjects with normal (NGT) or impaired (IGT) glucose tolerance and Type 2 diabetes mellitus (T2DM; n = 10-14 per group) ingested a liquid mixed meal plus acetaminophen (ACM; to assess gastric emptying) at time zero. On separate occasions, a primed-constant intravenous infusion of vehicle or Xen at 4 (Lo-Xen) or 12 (Hi-Xen) pmol · kg(-1) · min(-1) was administered from zero until 300 min. Some subjects with NGT received 30- and 90-min Hi-Xen infusions. Plasma ACM, glucose, insulin, C-peptide, glucagon, Xen, GIP, and glucagon-like peptide-1 (GLP-1) levels were measured and ISRs calculated. Areas under the curves were compared for treatment effects. Infusion with Hi-Xen, but not Lo-Xen, similarly delayed gastric emptying and reduced postprandial glucose levels in all groups. Infusions for 90 or 300 min, but not 30 min, were equally effective. Hi-Xen reduced plasma GLP-1, but not GIP, levels without altering the insulin secretory response to glucose. Intense staining for Xen receptors was detected on PGP9.5-positive nerve fibers in the longitudinal muscle of the human stomach. Thus Xen reduces gastric emptying in humans with and without T2DM, probably via a neural relay. Moreover, endogenous GLP-1 may not be a major enhancer of insulin secretion in healthy humans under physiological conditions.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Vaciamiento Gástrico/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Neurotensina/uso terapéutico , Periodo Posprandial , Adulto , Biomarcadores/sangre , Glucemia/metabolismo , Péptido C/sangre , Estudios Cruzados , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/fisiopatología , Esquema de Medicación , Femenino , Glucagón/sangre , Péptido 1 Similar al Glucagón/sangre , Humanos , Hipoglucemiantes/administración & dosificación , Infusiones Intravenosas , Insulina/sangre , Masculino , Persona de Mediana Edad , Missouri , Neurotensina/administración & dosificación , Receptores de Neurotensina/efectos de los fármacos , Receptores de Neurotensina/metabolismo , Factores de Tiempo , Resultado del Tratamiento
4.
Biotechnol Appl Biochem ; 57(4): 127-38, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21054278

RESUMEN

Recombinant GST (glutathione transferase) proteins are widely used as immunogens to generate polyclonal antibodies. Advantages of using GST proteins include: commercially available cloning vectors, vast literature for protein expression in Escherichia coli, the ease of protein purification, immunogen can be used as an ELISA standard and GST can be removed in some systems. However, there are disadvantages: GST oligomerization, inclusion body formation and target protein insolubility after GST removal. Perhaps the most detrimental is the significant generation of anti-GST antibodies by the host animal. A two-column procedure using a glutathione-GST column and a glutathione-(GST-protein) column can yield affinity-purified anti-(GST-protein) polyclonal antibody. Several passes over the first column are often required, though, to completely extract the anti-GST antibodies from the immune sera. We reasoned that knowledge of the target protein linear epitope(s) would allow construction of a peptide affinity resin for a single-pass 'one and done' purification termed ETRAP (efficient trapping and purification). In the present paper, we describe our efforts and present data on rabbits and sheep immunized with GST proteins having target protein molecular masses of ~8, 21 and 33 kDa. The titre and purity of the target antibodies using the ETRAP protocol were comparable to the more laborious multi-column purifications but with a considerable saving in time.


Asunto(s)
Anticuerpos/sangre , Anticuerpos/aislamiento & purificación , Cromatografía de Afinidad/métodos , Glutatión Transferasa/sangre , Sueros Inmunes/química , Animales , Anticuerpos/inmunología , Clonación Molecular , Epítopos/inmunología , Escherichia coli/genética , Glutatión Transferasa/inmunología , Inmunoensayo , Conejos , Proteínas Recombinantes de Fusión/sangre , Proteínas Recombinantes de Fusión/inmunología , Ovinos
5.
J Biol Chem ; 285(26): 19842-53, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20421298

RESUMEN

The intestinal peptides GLP-1 and GIP potentiate glucose-mediated insulin release. Agents that increase GLP-1 action are effective therapies in type 2 diabetes mellitus (T2DM). However, GIP action is blunted in T2DM, and GIP-based therapies have not been developed. Thus, it is important to increase our understanding of the mechanisms of GIP action. We developed mice lacking GIP-producing K cells. Like humans with T2DM, "GIP/DT" animals exhibited a normal insulin secretory response to exogenous GLP-1 but a blunted response to GIP. Pharmacologic doses of xenin-25, another peptide produced by K cells, restored the GIP-mediated insulin secretory response and reduced hyperglycemia in GIP/DT mice. Xenin-25 alone had no effect. Studies with islets, insulin-producing cell lines, and perfused pancreata indicated xenin-25 does not enhance GIP-mediated insulin release by acting directly on the beta-cell. The in vivo effects of xenin-25 to potentiate insulin release were inhibited by atropine sulfate and atropine methyl bromide but not by hexamethonium. Consistent with this, carbachol potentiated GIP-mediated insulin release from in situ perfused pancreata of GIP/DT mice. In vivo, xenin-25 did not activate c-fos expression in the hind brain or paraventricular nucleus of the hypothalamus indicating that central nervous system activation is not required. These data suggest that xenin-25 potentiates GIP-mediated insulin release by activating non-ganglionic cholinergic neurons that innervate the islets, presumably part of an enteric-neuronal-pancreatic pathway. Xenin-25, or molecules that increase acetylcholine receptor signaling in beta-cells, may represent a novel approach to overcome GIP resistance and therefore treat humans with T2DM.


Asunto(s)
Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Glucosa/farmacología , Neurotensina/farmacología , Animales , Glucemia/metabolismo , Western Blotting , Carbacol/farmacología , Línea Celular Tumoral , Agonistas Colinérgicos/farmacología , Sinergismo Farmacológico , Ensayo de Inmunoadsorción Enzimática , Ayuno/sangre , Femenino , Polipéptido Inhibidor Gástrico/genética , Polipéptido Inhibidor Gástrico/farmacología , Humanos , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurotensina/sangre , Páncreas/efectos de los fármacos , Páncreas/metabolismo
6.
Biochim Biophys Acta ; 1773(3): 321-9, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17140678

RESUMEN

We utilized the budding yeast Saccharomyces cerevisiae as a model to systematically explore physiological roles for yeast and mammalian aldo-keto reductases. Six open reading frames encoding putative aldo-keto reductases were identified when the yeast genome was queried against the sequence for human aldose reductase, the prototypical mammalian aldo-keto reductase. Recombinant proteins produced from five of these yeast open reading frames demonstrated NADPH-dependent reductase activity with a variety of aldehyde and ketone substrates. A triple aldo-keto reductase null mutant strain demonstrated a glucose-dependent heat shock phenotype which could be rescued by ectopic expression of human aldose reductase. Catalytically-inactive mutants of human or yeast aldo-keto reductases failed to effect a rescue of the heat shock phenotype, suggesting that the phenotype results from either an accumulation of one or more unmetabolized aldo-keto reductase substrates or a synthetic deficiency of aldo-keto reductase products generated in response to heat shock stress. These results suggest that multiple aldo-keto reductases fulfill functionally redundant roles in the stress response in yeast.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Saccharomyces cerevisiae/enzimología , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/aislamiento & purificación , Aldehído Reductasa , Aldo-Ceto Reductasas , Eliminación de Gen , Regulación Enzimológica de la Expresión Génica , Regulación Fúngica de la Expresión Génica , Concentración de Iones de Hidrógeno , Cinética , Mutación , Fenotipo , Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA