Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Microorganisms ; 11(2)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36838333

RESUMEN

Emerging variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) show immune evasion of vaccine-derived immunity, highlighting the need for better clinical immunogenicity biomarkers. To address this need, an enzyme-linked immunosorbent assay-based, human angiotensin-converting enzyme 2 (hACE2) binding inhibition assay was developed to measure antibodies against the ancestral strain of SARS-CoV-2 and was validated for precision, specificity, linearity, and other parameters. This assay measures the inhibition of SARS-CoV-2 spike (S) protein binding to the receptor, hACE2, by serum from vaccine clinical trials. Inter- and intra-assay precision, specificity, linearity, lower limit of quantitation, and assay robustness parameters successfully met the acceptance criteria. Heme and lipid matrix effects showed minimal interference on the assay. Samples were stable for testing in the assay even with 8 freeze/thaws and up to 24 months in -80 °C storage. The assay was also adapted for variants (Delta and Omicron BA.1/BA.5), with similar validation results. The hACE2 assay showed significant correlation with anti-recombinant S immunoglobulin G levels and neutralizing antibody titers. This assay provides a rapid, high-throughput option to evaluate vaccine immunogenicity. Along with other clinical biomarkers, it can provide valuable insights into immune evasion and correlates of protection and enable vaccine development against emerging COVID-19 variants.

2.
Free Radic Biol Med ; 42(1): 90-105, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17157196

RESUMEN

Poly(ADP-ribose) is synthesized from nicotinamide adenine dinucleotide (NAD) by poly(ADP-ribose) polymerase 1 (PARP-1) and degraded by poly(ADP-ribose) glycohydrolase (PARG). The aim of the present study was to examine the role of PARG in the development of experimental colitis. To address this question, we used an experimental model of colitis, induced by dinitrobenzene sulfonic acid (DNBS). Mice lacking the functional 110-kDa isoform of PARG (PARG(110)KO mice) were resistant to colon injury induced by DNBS. The mucosa of colon tissues showed reduction of myeloperoxidase activity and attenuated staining for intercellular adhesion molecule 1 and vascular cell adhesion molecule 1. Moreover, overproduction of proinflammatory factors TNF-alpha and IL-1beta and activation of cell death signaling pathway, i.e., the FAS ligand, were inhibited in these mutant mice. Finally pharmacological treatment of WT mice with GPI 16552 and 18214, two novel PARG inhibitors, showed a significant protective effect in DNBS-induced colitis. These genetic and pharmacological studies demonstrate that PARG modulates the inflammatory response and tissue injury events associated with colitis and PARG may be considered as a novel target for pharmacological intervention for the pathogenesis.


Asunto(s)
Colitis/terapia , Glicósido Hidrolasas/fisiología , Enfermedades Inflamatorias del Intestino/terapia , Animales , Bencenosulfonatos/toxicidad , Muerte Celular , Colitis/inducido químicamente , Colitis/enzimología , Modelos Animales de Enfermedad , Proteína Ligando Fas/metabolismo , Glicósido Hidrolasas/antagonistas & inhibidores , Glicósido Hidrolasas/genética , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/enzimología , Interleucina-1beta/metabolismo , Ratones , Peroxidasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
3.
Acta Biochim Pol ; 54(1): 63-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17311110

RESUMEN

Sirtuins (type III histone deacetylases) are an important member of a group of enzymes that modify chromatin conformation. We investigated the role of sirtuin inhibitor, GPI 19015, in double strand break (DSB) repair in CHO-K1 wt and xrs-6 mutant cells. The latter is defective in DNA-dependent protein kinase (DNA-PK)-mediated non-homologous end-joining (D-NHEJ). DSB were estimated by the neutral comet assay and histone gammaH2AX foci formation. We observed a weaker effect of GPI 19015 treatment on the repair kinetics in CHO wt cells than in xrs6. In the latter cells the increase in DNA repair rate was most pronounced in G1 phase and practically absent in S and G2 cell cycle phases. The decrease in the number of histone gammaH2AX foci was faster in xrs6 than in CHO-K1 cells. The altered repair rate did not affect survival of X-irradiated cells. Since in G1 xrs6 cells DNA-PK-dependent non-homologous end-joining, D-NHEJ, does not operate, these results indicate that inhibition of sirtuins modulates DNA-PK-independent (backup) non-homologous end-joining, B-NHEJ, to a greater extent than the other DSB repair system, D-NHEJ.


Asunto(s)
Daño del ADN , Reparación del ADN , Sirtuinas/antagonistas & inhibidores , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Histona Desacetilasa 2 , Inhibidores de Histona Desacetilasas , Cinética , Mutación , Proteínas Represoras/antagonistas & inhibidores
4.
Brain Res ; 978(1-2): 99-103, 2003 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-12834903

RESUMEN

Poly(ADP-ribose) is synthesized from nicotinamide adenine dinucleotide (NAD(+)) by poly(ADP-ribose) polymerase (PARP) and degraded by poly(ADP-ribose) glycohydrolase (PARG). Overactivation of the poly(ADP-ribose) pathway increases nicotinamide and decreases cellular NAD(+)/ATP, which leads to cell death. Blocking poly(ADP-ribose) metabolism by inactivating PARP has been shown to reduce ischemia injury. We investigated whether disrupting the poly(ADP-ribose) cycle by PARG inhibition could achieve similar protection. We demonstrate that either pre- or post-ischemia treatment with 40 mg/kg of N-bis-(3-phenyl-propyl)9-oxo-fluorene-2,7-diamide, a novel PARG inhibitor, significantly reduces brain infarct volumes by 40-53% in a rat model of focal cerebral ischemia. Our result provides the first evidence that PARG inhibitors can ameliorate ischemic brain damage in vivo, in support of PARG as a new therapeutic target for treating ischemia injury.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Infarto Cerebral/tratamiento farmacológico , Diamida/análogos & derivados , Diamida/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Glicósido Hidrolasas/antagonistas & inhibidores , Animales , Encéfalo/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Infarto Cerebral/metabolismo , Infarto Cerebral/patología , Colorantes/farmacocinética , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratas , Ratas Sprague-Dawley , Reperfusión/métodos , Sales de Tetrazolio/farmacocinética , Factores de Tiempo
5.
J Pharmacol Exp Ther ; 319(1): 127-38, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16825529

RESUMEN

The aim of the present study was to examine the role of poly-(ADP-ribose) glycohydrolase (PARG) on the modulation of the inflammatory response and tissue injury associated with neurotrauma. Spinal cord trauma was induced in wild-type (WT) mice by the application of vascular clips (force of 24 g) to the dura via a two-level T(6) to T(7) laminectomy. Spinal cord injury in WT mice resulted in severe trauma characterized by edema, neutrophil infiltration, and cytokine production followed by recruitment of other inflammatory cells, production of a range of inflammation mediators, tissue damage, apoptosis, and disease. The genetic disruption of the PARG gene in mice or the pharmacological inhibition of PARG with GPI 16552 [N-bis-(3-phenyl-propyl)9-oxo-fluorene-2,7-diamide] (40 mg/kg i.p. bolus), a novel and potent PARG inhibitor, significantly reduced the degree of spinal cord inflammation and tissue injury (histological score), neutrophil infiltration, cytokine production (tumor necrosis factor-alpha and interleukin-1beta), and apoptosis. In a separate experiment, we have clearly demonstrated that PARG inhibition significantly ameliorated the recovery of limb function. Taken together, our results indicate that PARG activity modulates the inflammatory response and tissue injury events associated with spinal cord trauma and participate in target organ damage under these conditions.


Asunto(s)
Glicósido Hidrolasas/fisiología , Inflamación/etiología , Traumatismos de la Médula Espinal/enzimología , Animales , Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Fluorenos/uso terapéutico , Glicósido Hidrolasas/antagonistas & inhibidores , Interleucina-1/biosíntesis , Masculino , Ratones , Infiltración Neutrófila/efectos de los fármacos , Peroxidasa/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Proteína X Asociada a bcl-2/análisis
6.
J Biol Chem ; 278(2): 745-50, 2003 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-12424231

RESUMEN

Tumor suppressor p53 exhibits an enigmatic phenotype in cells exposed to electrophilic, cyclopentenone prostaglandins of the A and J series. Namely, cells harboring a wild-type p53 gene accumulate p53 protein that is conformationally and functionally impaired. This occurs via an unknown molecular mechanism. We report that electrophilic cyclopentenone prostaglandins covalently modify and inhibit thioredoxin reductase, a selenoprotein that governs p53 and other redox-sensitive transcription factors. This mechanism accounts fully for the unusual p53 phenotype in cells exposed to electrophilic prostaglandins. Based on this mechanism we derived, tested, and affirmed several predictions regarding the kinetics of p53 inactivation; the protective effects of selenium; the structure-activity relationships for inhibition of thioredoxin reductase and impairment of p53 by electrophilic lipids; the susceptibility of hypoxia-inducible factor to inactivation by electrophilic lipids; and the equivalence of chemical inactivation of p53 to deletion of a p53 allele. Chemical precepts dictate that other electrophilic agents should also inhibit thioredoxin reductase and impair its governance of redox-sensitive proteins. Our results provide a novel framework to understand how endogenous and exogenous electrophiles might participate in carcinogenesis; how selenoproteins and selenium might confer protection against cancer; how certain tumors might acquire their paradoxical p53 phenotype; and how chronic inflammation might heighten the risk for cancer.


Asunto(s)
Aldehídos/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Prostaglandinas A/farmacología , Proteínas/antagonistas & inhibidores , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Factores de Transcripción , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Apoptosis , Línea Celular , Etopósido/farmacología , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Oxidación-Reducción , Conformación Proteica , Proteínas/fisiología , Proteínas Represoras/farmacología , Selenoproteínas , Reductasa de Tiorredoxina-Disulfuro/fisiología , Proteína p53 Supresora de Tumor/química
7.
Crit Care Med ; 32(6): 1365-74, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15187521

RESUMEN

OBJECTIVE: Poly(ADP-ribose) is synthesized from nicotinamide adenine dinucleotide by poly(ADP-ribose) polymerase (PARP) and degraded by poly(ADP-ribose) glycohydrolase (PARG). The activation of the PARP/PARG pathway has been found in a variety of animal models of diseases, including septic shock-like syndrome. We have previously demonstrated that PARP inhibition by 3-ami-nobenzamide or GPI 6150 ameliorates multiple organ dysfunctions induced by zymosan. In the present study, we investigated whether similar effect could be achieved through PARG inhibition to break the cycle of poly(ADP-ribose) turnaround. DESIGN: Experimental study. SETTING: University laboratory. SUBJECTS: Male CD mice (20-22 g). INTERVENTIONS: We tested the effects of GPI 18214 (40 mg/kg intraperitoneally bolus), a novel and potent PARG inhibitor, at 1 and 6 hr after zymosan (500 mg/kg, administered intraperitoneally as a suspension in saline) on the development of septic shock-like syndrome in mice. Organ failure and systemic inflammation in mice were assessed 18 hrs after administration of zymosan and/or GPI 18214 and monitored for 12 days (for loss of body weight and mortality). MEASUREMENTS AND MAIN RESULTS: At 18 hrs after zymosan administration, we found a significant increase of peritoneal exudates, leukocyte infiltration in peritoneal cavity as well as an infiltration of neutrophils in lung and ileum tissues and subsequent lipid peroxidation, and increased production of plasma tumor necrosis factor-alpha and interleukin-1 beta. Furthermore, zymosan administration induced significant liver, lung, pancreas, intestine, and kidney dysfunction as well as a systemic toxicity and significant loss of body weight. At the end of observation period (12 days), 90% of zymosan-treated mice were dead. GPI 18214 (40 mg/kg intraperitoneally, 1 and 6 hrs after zymosan) treatment significantly reduced peritoneal exudates, inflammatory cell infiltration, and organ injury and mortality rate in zymosan-treated mice. CONCLUSIONS: This study supports early studies that show efficacy from blocking the poly(ADP-ribose) pathway in septic shock-like syndrome model. It provides evidence that GPI 18214, a PARG inhibitor, attenuates the degree of zymosan-induced nonseptic shock in mice, suggesting that PARG may be an alternative therapeutic target for shock treatment.


Asunto(s)
Amidas/uso terapéutico , Glicósido Hidrolasas/antagonistas & inhibidores , Choque Séptico/prevención & control , Taninos/uso terapéutico , Enfermedad Aguda , Amidas/administración & dosificación , Animales , Interleucina-1/biosíntesis , Masculino , Malondialdehído/análisis , Ratones , Ratones Endogámicos , Insuficiencia Multiorgánica/tratamiento farmacológico , Peritonitis/inducido químicamente , Peroxidasa/metabolismo , Choque Séptico/inducido químicamente , Choque Séptico/tratamiento farmacológico , Choque Séptico/patología , Taninos/administración & dosificación , Factor de Necrosis Tumoral alfa/biosíntesis , Zimosan
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA