Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Chembiochem ; 22(24): 3469-3476, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34569136

RESUMEN

The dynamic interplay of post-translational modifications (PTMs) in chromatin provides a communication system for the regulation of gene expression. An increasing number of studies have highlighted the role that such crosstalk between PTMs plays in chromatin recognition. In this study, (bio)chemical and structural approaches were applied to specifically probe the impact of acetylation of Lys18 in the histone H3 tail peptide on peptide recognition by the protein methyltransferase coactivator-associated arginine methyltransferase 1 (CARM1). Peptidomimetics that recapitulate the transition state of protein arginine N-methyltransferases, were designed based on the H3 peptide wherein the target Arg17 was flanked by either a free or an acetylated lysine. Structural studies with these peptidomimetics and the catalytic domain of CARM1 provide new insights into the binding of the H3 peptide within the enzyme active site. While the co-crystal structures reveal that lysine acetylation results in minor conformational differences for both CARM1 and the H3 peptide, acetylation of Lys18 does lead to additional interactions (Van der Waals and hydrogen bonding) and likely reduces the cost of desolvation upon binding, resulting in increased affinity. Informed by these findings a series of smaller peptidomimetics were also prepared and found to maintain potent and selective CARM1 inhibition. These findings provide new insights both into the mechanism of crosstalk between arginine methylation and lysine acetylation as well as towards the development of peptidomimetic CARM1 inhibitors.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Lisina/antagonistas & inhibidores , Peptidomiméticos/farmacología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Acetilación , Animales , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Lisina/metabolismo , Ratones , Modelos Moleculares , Peptidomiméticos/síntesis química , Peptidomiméticos/química , Conformación Proteica , Proteína-Arginina N-Metiltransferasas/metabolismo , Especificidad por Sustrato
2.
Bioorg Med Chem Lett ; 28(2): 167-169, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29198865

RESUMEN

Lysine-specific demethylase 1 (LSD1) is a flavin-dependent enzyme that removes methyl groups from mono- or dimethylated lysine residues at the fourth position of histone H3. We have previously reported several histone H3 peptides containing an LSD1 inactivator motif at Lys-4. In this study, histone H3 peptides having a trans-2-phenylcyclopropylamine (PCPA), a 2,5-dihydro-1H-pyrrole, and a 1,2,3,6-tetrahydropyridine moiety at Lys-4 were prepared along with related compounds possessing a shorter side chain at the fourth position. Enzymatic assays showed that PCPA peptides containing a longer side chain, which can react with FAD in the active site, are potent LSD1-selective inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Histona Demetilasas/antagonistas & inhibidores , Histonas/farmacología , Lisina/antagonistas & inhibidores , Péptidos/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Histona Demetilasas/metabolismo , Histonas/química , Humanos , Lisina/metabolismo , Estructura Molecular , Péptidos/química , Relación Estructura-Actividad
3.
Bioorg Med Chem ; 24(21): 5340-5352, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27622749

RESUMEN

γ-Glutamyl transpeptidase (GGT, EC 2.3.2.2) that catalyzes the hydrolysis and transpeptidation of glutathione and its S-conjugates is involved in a number of physiological and pathological processes through glutathione metabolism and is an attractive pharmaceutical target. We report here the evaluation of a phosphonate-based irreversible inhibitor, 2-amino-4-{[3-(carboxymethyl)phenoxy](methoyl)phosphoryl}butanoic acid (GGsTop) and its analogues as a mechanism-based inhibitor of human GGT. GGsTop is a stable compound, but inactivated the human enzyme significantly faster than the other phosphonates, and importantly did not inhibit a glutamine amidotransferase. The structure-activity relationships, X-ray crystallography with Escherichia coli GGT, sequence alignment and site-directed mutagenesis of human GGT revealed a critical electrostatic interaction between the terminal carboxylate of GGsTop and the active-site residue Lys562 of human GGT for potent inhibition. GGsTop showed no cytotoxicity toward human fibroblasts and hepatic stellate cells up to 1mM. GGsTop serves as a non-toxic, selective and highly potent irreversible GGT inhibitor that could be used for various in vivo as well as in vitro biochemical studies.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Escherichia coli/enzimología , Lisina/antagonistas & inhibidores , Organofosfonatos/farmacología , gamma-Glutamiltransferasa/antagonistas & inhibidores , Dominio Catalítico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Lisina/metabolismo , Modelos Moleculares , Estructura Molecular , Organofosfonatos/síntesis química , Organofosfonatos/química , Electricidad Estática , Relación Estructura-Actividad , gamma-Glutamiltransferasa/química , gamma-Glutamiltransferasa/metabolismo
4.
J Biol Regul Homeost Agents ; 30(3): 713-726, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27655488

RESUMEN

Osteoporosis is a metabolic multifaceted disorder, characterized by insufficient bone strength. It has been recently shown that advanced glycation end products (AGEs) play a role in senile osteoporosis, through bone cell impairment and altered biomechanical properties. Pentosidine (PENT), a wellcharacterized AGE, is also considered a biomarker of bone fracture. Adequate responses to various hormones, such as 1,25-dihydroxyvitamin D3, are prerequisites for optimal osteoblasts functioning. Vitamin K2 is known to enhance in vitro and in vitro vitamin D-induced bone formation. The aim of the study was to assess the effects of Vitamins D3 and K2 and PENT on in vitro osteoblast activity, to convey a possible translational clinical message. Ex vivo human osteoblasts cultured, for 3 weeks, with vitamin D3 and vitamin K2 were exposed to PENT, a well-known advanced glycoxidation end product for the last 72 hours. Experiments with PENT alone were also carried out. Gene expression of specific markers of bone osteoblast maturation [alkaline phosphatase, ALP; collagen I, COL Iα1; and osteocalcin (bone-Gla-protein) BGP] was measured, together with the receptor activator of nuclear factor kappa-B ligand/osteoproteregin (RANKL/OPG) ratio to assess bone remodeling. Expression of RAGE, a well-characterized receptor of AGEs, was also assessed. PENT+vitamins slightly inhibited ALP secretion while not affecting gene expression, indicating hampered osteoblast functional activity. PENT+vitamins up-regulated collagen gene expression, while protein secretion was unchanged. Intracellular collagen levels were partially decreased, and a significant reduction in BGP gene expression and intracellular protein concentration were both reported after PENT exposure. The RANKL/OPG ratio was increased, favouring bone reabsorption. RAGE gene expression significantly decreased. These results were confirmed by a lower mineralization rate. We provided in vitro evidence that glycoxidation might interfere with the maturation of osteoblasts, leading to morphological modifications, cellular malfunctioning, and inhibition of the calcification process. However, these processes may be all partially counterbalanced by vitamins D3 and K2. Therefore, detrimental AGE accumulation in bone might be attenuated and/or reversed by the presence or supplementation of vitamins D3 and K2.


Asunto(s)
Arginina/análogos & derivados , Colecalciferol/farmacología , Lisina/análogos & derivados , Osteoblastos/efectos de los fármacos , Vitamina K 2/farmacología , Fosfatasa Alcalina/biosíntesis , Fosfatasa Alcalina/genética , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Arginina/antagonistas & inhibidores , Arginina/toxicidad , Remodelación Ósea/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lisina/antagonistas & inhibidores , Lisina/toxicidad , Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Proteínas Quinasas Activadas por Mitógenos/genética , Osteoblastos/metabolismo , Osteocalcina/biosíntesis , Osteocalcina/genética , Osteogénesis/efectos de los fármacos , Osteoprotegerina/biosíntesis , Osteoprotegerina/genética , Ligando RANK/biosíntesis , Ligando RANK/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética
5.
Acta Pharmacol Sin ; 37(8): 1054-62, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27264315

RESUMEN

AIM: The aim of this study was to examine the activation of neuronal Kv7/KCNQ channels by a novel modified Kv7 opener QO58-lysine and to test the anti-nociceptive effects of QO58-lysine on inflammatory pain in rodent models. METHODS: Assays including whole-cell patch clamp recordings, HPLC, and in vivo pain behavioral evaluations were employed. RESULTS: QO58-lysine caused instant activation of Kv7.2/7.3 currents, and increasing the dose of QO58-lysine resulted in a dose-dependent activation of Kv7.2/Kv7.3 currents with an EC50 of 1.2±0.2 µmol/L. QO58-lysine caused a leftward shift of the voltage-dependent activation of Kv7.2/Kv7.3 to a hyperpolarized potential at V1/2=-54.4±2.5 mV from V1/2=-26.0±0.6 mV. The half-life in plasma (t1/2) was derived as 2.9, 2.7, and 3.0 h for doses of 12.5, 25, and 50 mg/kg, respectively. The absolute bioavailabilities for the three doses (12.5, 25, and 50 mg/kg) of QO58-lysine (po) were determined as 13.7%, 24.3%, and 39.3%, respectively. QO58-lysine caused a concentration-dependent reduction in the licking times during phase II pain induced by the injection of formalin into the mouse hindpaw. In the Complete Freund's adjuvant (CFA)-induced inflammatory pain model in rats, oral or intraperitoneal administration of QO58-lysine resulted in a dose-dependent increase in the paw withdrawal threshold, and the anti-nociceptive effect on mechanical allodynia could be reversed by the channel-specific blocker XE991 (3 mg/kg). CONCLUSION: Taken together, our findings show that a modified QO58 compound (QO58-lysine) can specifically activate Kv7.2/7.3/M-channels. Oral or intraperitoneal administration of QO58-lysine, which has improved bioavailability and a half-life of approximately 3 h in plasma, can reverse inflammatory pain in rodent animal models.


Asunto(s)
Canales de Potasio KCNQ/agonistas , Lisina/farmacología , Dimensión del Dolor/efectos de los fármacos , Animales , Antracenos/farmacología , Disponibilidad Biológica , Carbamatos/farmacología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Lisina/antagonistas & inhibidores , Lisina/farmacocinética , Masculino , Potenciales de la Membrana/efectos de los fármacos , Fenilendiaminas/farmacología , Ratas
6.
Am J Physiol Endocrinol Metab ; 309(10): E829-39, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26394662

RESUMEN

Nε-(carboxymethyl) lysine-conjugated bovine serum albumin (CML-BSA) is a major component of advanced glycation end products (AGEs). We hypothesised that AGEs reduce insulin secretion from pancreatic ß-cells by damaging mitochondrial functions and inducing mitophagy. Mitochondrial morphology and the occurrence of autophagy were examined in pancreatic islets of diabetic db/db mice and in the cultured CML-BSA-treated insulinoma cell line RIN-m5F. In addition, the effects of α-lipoic acid (ALA) on mitochondria in AGE-damaged tissues were evaluated. The diabetic db/db mouse exhibited an increase in the number of autophagosomes in damaged mitochondria and receptor for AGEs (RAGE). Treatment of db/db mice with ALA for 12 wk increased the number of mitochondria with well-organized cristae and fewer autophagosomes. Treatment of RIN-m5F cells with CML-BSA increased the level of RAGE protein and autophagosome formation, caused mitochondrial dysfunction, and decreased insulin secretion. CML-BSA also reduced mitochondrial membrane potential and ATP production, increased ROS and lipid peroxide production, and caused mitochondrial DNA deletions. Elevated fission protein dynamin-related protein 1 (Drp1) level and mitochondrial fragmentation demonstrated the unbalance of mitochondrial fusion and fission in CML-BSA-treated cells. Additionally, increased levels of Parkin and PTEN-induced putative kinase 1 protein suggest that fragmented mitochondria were associated with increased mitophagic activity, and ALA attenuated the CML-BSA-induced mitophage formation. Our study demonstrated that CML-BSA induced mitochondrial dysfunction and mitophagy in pancreatic ß-cells. The findings from this study suggest that increased concentration of AGEs may damage ß-cells and reduce insulin secretion.


Asunto(s)
Diabetes Mellitus/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Lisina/análogos & derivados , Dinámicas Mitocondriales , Mitofagia , Animales , Antioxidantes/metabolismo , Antioxidantes/uso terapéutico , Biomarcadores/metabolismo , Línea Celular Tumoral , Diabetes Mellitus/dietoterapia , Diabetes Mellitus/patología , Suplementos Dietéticos , Regulación hacia Abajo/efectos de los fármacos , Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Productos Finales de Glicación Avanzada/farmacología , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/ultraestructura , Lisina/antagonistas & inhibidores , Lisina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Dinámicas Mitocondriales/efectos de los fármacos , Mitofagia/efectos de los fármacos , Oxidantes/antagonistas & inhibidores , Oxidantes/farmacología , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/ultraestructura , Ratas , Receptor para Productos Finales de Glicación Avanzada/agonistas , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Albúmina Sérica Bovina/antagonistas & inhibidores , Albúmina Sérica Bovina/farmacología , Ácido Tióctico/metabolismo , Ácido Tióctico/uso terapéutico
7.
Nat Chem Biol ; 9(3): 184-91, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23292653

RESUMEN

We describe the discovery of UNC1215, a potent and selective chemical probe for the methyllysine (Kme) reading function of L3MBTL3, a member of the malignant brain tumor (MBT) family of chromatin-interacting transcriptional repressors. UNC1215 binds L3MBTL3 with a K(d) of 120 nM, competitively displacing mono- or dimethyllysine-containing peptides, and is greater than 50-fold more potent toward L3MBTL3 than other members of the MBT family while also demonstrating selectivity against more than 200 other reader domains examined. X-ray crystallography identified a unique 2:2 polyvalent mode of interaction between UNC1215 and L3MBTL3. In cells, UNC1215 is nontoxic and directly binds L3MBTL3 via the Kme-binding pocket of the MBT domains. UNC1215 increases the cellular mobility of GFP-L3MBTL3 fusion proteins, and point mutants that disrupt the Kme-binding function of GFP-L3MBTL3 phenocopy the effects of UNC1215 on localization. Finally, UNC1215 was used to reveal a new Kme-dependent interaction of L3MBTL3 with BCLAF1, a protein implicated in DNA damage repair and apoptosis.


Asunto(s)
Benzamidas/farmacología , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Descubrimiento de Drogas , Lisina/análogos & derivados , Sondas Moleculares/farmacología , Piperidinas/farmacología , Benzamidas/química , Benzamidas/metabolismo , Unión Competitiva/efectos de los fármacos , Cristalografía por Rayos X , Proteínas de Unión al ADN/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Lisina/antagonistas & inhibidores , Lisina/química , Lisina/metabolismo , Modelos Moleculares , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Estructura Molecular , Piperidinas/química , Piperidinas/metabolismo , Estructura Terciaria de Proteína , Proteínas Represoras/metabolismo , Relación Estructura-Actividad , Proteínas Supresoras de Tumor/metabolismo
8.
Bioorg Med Chem Lett ; 25(17): 3481-7, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26220157

RESUMEN

Built upon our lead pan-SIRT1/2/3 tripeptidic inhibitors that contain the catalytic mechanism-based sirtuin inhibitory warhead N(ε)-thioacetyl-lysine, three of their analogs (i.e., 7, 9, and 19) were discovered in the current study to exhibit a significantly enhanced SIRT3 inhibitory selectivity while maintaining the SIRT3 inhibitory potency. These compounds represent novel lead compounds for developing more potent and selective SIRT3 inhibitors of the catalytic mechanism-based type.


Asunto(s)
Lisina/análogos & derivados , Sirtuina 3/genética , Sirtuina 3/metabolismo , Sirtuinas/química , Catálisis , Humanos , Lisina/antagonistas & inhibidores , Modelos Moleculares
9.
Lipids Health Dis ; 14: 161, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26645467

RESUMEN

BACKGROUND: In non-dialysis chronic kidney disease (CKD) patients with dyslipidemia, statin therapy is recommended to prevent cardiovascular complications. Dyslipidemia has been also shown to be an independent risk factor for the progression of CKD. However, it is still unclear whether statin therapy exerts an inhibitory effect on renal deterioration in CKD patients with dyslipidemia. The purpose of the present study was to examine possible therapeutic effects of statin add-on therapy on renal function as well as parameters of lipid and glucose metabolism, arterial stiffness and oxidative stress, in comparison to diet therapy, in CKD patients with dyslipidemia. METHODS: This study was a randomized, open-label, and parallel-group trial consisted of a 12-months treatment period in non-dialysis CKD patients with alubuminuria and dyslipidemia. Twenty eight patients were randomly assigned either to receive diet counseling alone (diet therapy group) or diet counseling plus pitavastatin (diet-plus-statin therapy group), to achieve the LDL-cholesterol (LDL-C) target of <100 mg/dl. RESULTS: The statin treatment by pitavastatin was well tolerated in all of the patients without any significant adverse events and the average dose of pitavastatin was 1.0 ± 0.0 mg daily after treatment. After the 12-months treatment period, LDL-C was significantly lower in the diet-plus-statin therapy group compared with the diet therapy group (diet vs diet-plus-statin: LDL-C, 126 ± 5 vs 83 ± 4 mg/dL, P < 0.001). On the other hand, the diet-plus-statin therapy did not significantly reduce albuminuria or delay the decline in eGFR compared with the diet therapy, and there was no relationship between the change in LDL-C and the change in eGFR or albuminuria. However, diet therapy as well as diet-plus-statin therapy exerted similar lowering effects on the pentosidine levels (diet therapy group, baseline vs 12 months: 40 ± 4 vs 24 ± 3 ng/mL, P = 0.001; diet-plus-statin therapy, 46 ± 7 vs 34 ± 6 ng/mL, P = 0.008). Furthermore, the results of multivariate regression analysis indicated that the change in pentosidine was a significant contributor to the change in eGFR (ß = -0.536, P = 0.011). CONCLUSIONS: Although statin add-on therapy did not show additive renal protective effects, the diet therapy as well as the diet-plus-statin therapy could contribute to the reduction in plasma pentosidine in CKD patients with albuminuria and dyslipidemia.


Asunto(s)
Albuminuria/tratamiento farmacológico , Anticolesterolemiantes/uso terapéutico , Dieta/métodos , Dislipidemias/tratamiento farmacológico , Quinolinas/uso terapéutico , Insuficiencia Renal Crónica/tratamiento farmacológico , Anciano , Albuminuria/sangre , Albuminuria/dietoterapia , Albuminuria/patología , Arginina/análogos & derivados , Arginina/antagonistas & inhibidores , Arginina/sangre , HDL-Colesterol/sangre , LDL-Colesterol/antagonistas & inhibidores , LDL-Colesterol/sangre , Dislipidemias/sangre , Dislipidemias/dietoterapia , Dislipidemias/patología , Femenino , Tasa de Filtración Glomerular , Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Productos Finales de Glicación Avanzada/sangre , Humanos , Lisina/análogos & derivados , Lisina/antagonistas & inhibidores , Lisina/sangre , Masculino , Persona de Mediana Edad , Estrés Oxidativo/efectos de los fármacos , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/dietoterapia , Insuficiencia Renal Crónica/patología , Triglicéridos/sangre , Rigidez Vascular/efectos de los fármacos
10.
Biochem J ; 442(1): 221-30, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22060242

RESUMEN

Non-enzymatic glycation is a complex series of reactions between reducing sugars and amino groups of proteins. Accumulation of AGEs (advanced glycation end-products) due to non-enzymatic glycation has been related to several diseases associated with aging and diabetes. The formation of AGEs is accelerated in hyperglycaemic conditions, which alters the structure and function of long-lived proteins, thereby contributing to long-term diabetic complications. The present study describes AGE inhibition and the mechanism of action of a new antiglycating agent, EA (ellagic acid), a flavonoid present in many dietary sources. Inhibition of AGE formation by EA was demonstrated with different proteins, namely eye lens TSP (total soluble protein), Hb (haemoglobin), lysozyme and BSA, using different glycating agents such as fructose, ribose and methylglyoxal by a set of complementary methods. These results suggest that the antiglycating action of EA seems to involve, apart from inhibition of a few fluorescent AGEs, predominantly inhibition of CEL [Nϵ-(carboxyethyl)lysine] through scavenging of the dicarbonyl compounds. Furthermore, MALDI-TOF-MS (matrix-assisted laser-desorption ionisation-time-of-flight MS) analysis confirms inhibition of the formation of CEL on lysozyme on in vitro glycation by EA. Prevention of glycation-mediated ß-sheet formation in Hb and lysozyme by EA confirm its antiglycating ability. Inhibition of glycosylated Hb formation in human blood under ex vivo high-glucose conditions signifies the physiological antiglycating potential of EA. We have also determined the effectiveness of EA against loss of eye lens transparency through inhibition of AGEs in the lens organ culture system. These findings establish the antiglycating potential of EA and its in vivo utility in controlling AGE-mediated diabetic pathologies.


Asunto(s)
Ácido Elágico/farmacología , Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Lisina/análogos & derivados , Animales , Cristalinas/química , Cristalinas/efectos de los fármacos , Complicaciones de la Diabetes/prevención & control , Gluconatos/química , Hemoglobina Glucada/biosíntesis , Productos Finales de Glicación Avanzada/inmunología , Humanos , Lactonas/química , Cristalino/efectos de los fármacos , Cristalino/metabolismo , Lisina/antagonistas & inhibidores , Masculino , Muramidasa/química , Muramidasa/efectos de los fármacos , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
11.
Planta Med ; 77(2): 196-204, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20717877

RESUMEN

Nonenzymatic formation of advanced glycation end products (AGEs) is accelerated under hyperglycemic conditions characteristic of type 2 diabetes mellitus and contributes to the development of vascular complications. As such, inhibition of AGE formation represents a potential therapeutic target for the prevention and treatment of diabetic complications. In the present study, ethanolic extracts of 17 medicinal plants were assessed for inhibitory effects on in vitro AGE formation through fluorometric and immunochemical detection of fluorescent AGEs and N(ε)-(carboxymethyl)lysine adducts of albumin (CML-BSA), respectively. Most extracts inhibited fluorescent AGE formation with IC (50) values ranging from 0.4 to 38.6 µg/mL and all extracts reduced CML-BSA formation but to differing degrees. Results obtained through both methods were highly correlated. Antiglycation activities were positively correlated with total phenolic content, free radical scavenging activity and reduction in malonyldiadehyde levels following oxidation of low-density lipoprotein, but negatively correlated with lag time to formation of conjugated dienes. Together, these results provide evidence that antioxidant phenolic metabolites mediate the antiglycation activity of our medicinal plant collection, a relationship that likely extends to other medicinal and food plants.


Asunto(s)
Antioxidantes/farmacología , Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Fenoles/farmacología , Extractos Vegetales/farmacología , Plantas Medicinales/química , Canadá , Depuradores de Radicales Libres/química , Productos Finales de Glicación Avanzada/química , Productos Finales de Glicación Avanzada/metabolismo , Lisina/análogos & derivados , Lisina/antagonistas & inhibidores , Lisina/química , Malondialdehído/química , Oxidación-Reducción , Extractos Vegetales/química , Factores de Tiempo
12.
Poult Sci ; 100(2): 1308-1318, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33518088

RESUMEN

Advanced glycation end products (AGE), compounds formed in meat at the advanced stage of Maillard reaction, are easily exposed to thermal processing. Improving cooking condition and adding antioxidants are 2 common ways for AGE reduction. The present work compared the inhibition of grape seed extract (GSE) on levels of free and protein-bound Nε-carboxymethyllysine (CML) and Nε-carboxyethyllysine (CEL) in chicken breast under deep-frying and air-frying conditions. Efficiency of 5 concentrations of GSE (0.0, 0.2, 0.5, 0.8, and 1.0 g/kg) in retarding oxidation, glyoxal (GO), methylglyoxal (MGO), lysine (Lys), Maillard reaction degree (A294, A420), and Shiff's base were tested. Results showed that 0.5 g/kg GSE before heating significantly (P < 0.05) reduced AGE in fried breast chicken, whereas excessive supplementation of GSE (0.8 and 1 g/kg) was reverse. Air frying was found significantly (P < 0.05) better than deep frying to reduce the precursor substances (GO, MGO, and Lys) of AGE. In conclusion, GSE-derived polyphenols exhibited different inhibitory effects on oxidation and glycosylation at different concentrations. We found that 0.5 g/kg of GSE combined with air frying was the best recommendation for inhibiting CML and CEL.


Asunto(s)
Culinaria/métodos , Extracto de Semillas de Uva/farmacología , Lisina/análogos & derivados , Carne/normas , Animales , Pollos , Productos Finales de Glicación Avanzada/análisis , Productos Finales de Glicación Avanzada/metabolismo , Glicosilación , Lisina/antagonistas & inhibidores , Lisina/metabolismo , Reacción de Maillard , Fibras Musculares Esqueléticas/citología , Oxidación-Reducción , Compuestos de Sulfhidrilo/análisis , Sustancias Reactivas al Ácido Tiobarbitúrico/análisis
13.
Eur J Med Chem ; 225: 113803, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34461505

RESUMEN

Human sirtuin 5 (SIRT5) plays pivotal roles in metabolic pathways and other biological processes, and is involved in several human diseases including cancer. Development of new potent and selective SIRT5 inhibitors is currently desirable to provide potential therapeutics for related diseases. Herein, we report a series of new 3-thioureidopropanoic acid derivatives, which were designed to mimic the binding features of SIRT5 glutaryl-lysine substrates. Structure-activity relationship studies revealed several compounds with low micromolar inhibitory activities to SIRT5. Computational and biochemical studies indicated that these compounds exhibited competitive SIRT5 inhibition with respect to the glutaryl-lysine substrate rather than nicotinamide adenine dinucleotide cofactor. Moreover, they showed high selectivity for SIRT5 over SIRT1-3 and 6 and could stabilize SIRT5 proteins as revealed by thermal shift analyses. This work provides an effective substrate-mimicking strategy for future inhibitor design, and offers new inhibitors to investigate their therapeutic potentials in SIRT5-associated disease models.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Lisina/antagonistas & inhibidores , Propionatos/farmacología , Sirtuinas/antagonistas & inhibidores , Tiourea/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Lisina/metabolismo , Estructura Molecular , Propionatos/síntesis química , Propionatos/química , Sirtuinas/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato , Tiourea/síntesis química , Tiourea/química
14.
Eur J Med Chem ; 225: 113786, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34464874

RESUMEN

The emergence of the C797S mutation in EGFR is a frequent mechanism of resistance to osimertinib in the treatment of non-small cell lung cancer (NSCLC). In the present work, we report the design, synthesis and biochemical characterization of UPR1444 (compound 11), a new sulfonyl fluoride derivative which potently and irreversibly inhibits EGFRL858R/T790M/C797S through the formation of a sulfonamide bond with the catalytic residue Lys745. Enzymatic assays show that compound 11 displayed an inhibitory activity on EGFRWT comparable to that of osimertinib, and it resulted more selective than the sulfonyl fluoride probe XO44, recently reported to inhibit a significant part of the kinome. Neither compound 11 nor XO44 inhibited EGFRdel19/T790M/C797S triple mutant. When tested in Ba/F3 cells expressing EGFRL858R/T790M/C797S, compound 11 resulted significantly more potent than osimertinib at inhibiting both EGFR autophosphorylation and proliferation, even if the inhibition of EGFR autophosphorylation by compound 11 in Ba/F3 cells was not long lasting.


Asunto(s)
Lisina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Ácidos Sulfínicos/farmacología , Animales , Biocatálisis , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Lisina/metabolismo , Ratones , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Ácidos Sulfínicos/síntesis química , Ácidos Sulfínicos/química
15.
J Cereb Blood Flow Metab ; 41(5): 1080-1090, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32615885

RESUMEN

In eukaryotes, the polyamine pathway generates spermidine that activates the hypusination of the translation factor eukaryotic initiation factor 5A (eIF5A). Hypusinated-eIF5A modulates translation, elongation, termination and mitochondrial function. Evidence in model organisms like drosophila suggests that targeting polyamines synthesis might be of interest against ischemia. However, the potential of targeting eIF5A hypusination in stroke, the major therapeutic challenge specific to ischemia, is currently unknown. Using in vitro models of ischemic-related stress, we documented that GC7, a specific inhibitor of a key enzyme in the eIF5A activation pathway, affords neuronal protection. We identified the preservation of mitochondrial function and thereby the prevention of toxic ROS generation as major processes of GC7 protection. To represent a thoughtful opportunity of clinical translation, we explored whether GC7 administration reduces the infarct volume and functional deficits in an in vivo transient focal cerebral ischemia (tFCI) model in mice. A single GC7 pre- or post-treatment significantly reduces the infarct volume post-stroke. Moreover, GC7-post-treatment significantly improves mouse performance in the rotarod and Morris water-maze, highlighting beneficial effects on motor and cognitive post-stroke deficits. Our results identify the targeting of the polyamine-eIF5A-hypusine axis as a new therapeutic opportunity and new paradigm of research in stroke and ischemic diseases.


Asunto(s)
Guanina/análogos & derivados , Lisina/análogos & derivados , Mitocondrias/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/antagonistas & inhibidores , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Accidente Cerebrovascular/terapia , Animales , Conducta Animal/efectos de los fármacos , Cognición/efectos de los fármacos , Guanina/administración & dosificación , Guanina/farmacología , Guanina/uso terapéutico , Inyecciones Intraperitoneales , Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/prevención & control , Lisina/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/ultraestructura , Modelos Animales , Neuroprotección/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Factores de Iniciación de Péptidos/efectos de los fármacos , Poliaminas/metabolismo , Proteínas de Unión al ARN/efectos de los fármacos , Especies Reactivas de Oxígeno/toxicidad , Accidente Cerebrovascular/metabolismo , Factor 5A Eucariótico de Iniciación de Traducción
16.
BMC Bioinformatics ; 11 Suppl 1: S53, 2010 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-20122228

RESUMEN

BACKGROUND: An explosive global spreading of multidrug resistant Mycobacterium tuberculosis (Mtb) is a catastrophe, which demands an urgent need to design or develop novel/potent antitubercular agents. The Lysine/DAP biosynthetic pathway is a promising target due its specific role in cell wall and amino acid biosynthesis. Here, we report identification of potential antitubercular candidates targeting Mtb dihydrodipicolinate synthase (DHDPS) enzyme of the pathway using virtual screening protocols. RESULTS: In the present study, we generated three sets of drug-like molecules in order to screen potential inhibitors against Mtb drug target DHDPS. The first set of compounds was a combinatorial library, which comprised analogues of pyruvate (substrate of DHDPS). The second set of compounds consisted of pyruvate-like molecules i.e. structurally similar to pyruvate, obtained using 3D flexible similarity search against NCI and PubChem database. The third set constituted 3847 anti-infective molecules obtained from PubChem. These compounds were subjected to Lipinski's rule of drug-like five filters. Finally, three sets of drug-like compounds i.e. 4088 pyruvate analogues, 2640 pyruvate-like molecules and 1750 anti-infective molecules were docked at the active site of Mtb DHDPS (PDB code: 1XXX used in the molecular docking calculations) to select inhibitors establishing favorable interactions. CONCLUSION: The above-mentioned virtual screening procedures helped in the identification of several potent candidates that possess inhibitory activity against Mtb DHDPS. Therefore, these novel scaffolds/candidates which could have the potential to inhibit Mtb DHDPS enzyme would represent promising starting points as lead compounds and certainly aid the experimental designing of antituberculars in lesser time.


Asunto(s)
Antituberculosos/química , Ácido Diaminopimélico/antagonistas & inhibidores , Lisina/antagonistas & inhibidores , Mycobacterium tuberculosis/metabolismo , Sitios de Unión , Dominio Catalítico , Bases de Datos Factuales , Ácido Diaminopimélico/metabolismo , Diseño de Fármacos , Hidroliasas/antagonistas & inhibidores , Hidroliasas/química , Hidroliasas/metabolismo , Lisina/metabolismo , Modelos Moleculares , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Ácido Pirúvico/metabolismo , Relación Estructura-Actividad , Tuberculosis Resistente a Múltiples Medicamentos
17.
Free Radic Res ; 52(11-12): 1387-1397, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30693839

RESUMEN

Nε-(carboxymethyl) lysine (CML) plays causal roles in diabetic complications. In the present study, we investigated whether CML-induced HIF-1α accumulation and epithelial-mesenchymal transition (EMT) in HK-2 renal proximal tubular epithelial cells. Treatment with CML-BSA increased reactive oxygen species (ROS) production reduced the mitochondrial membrane potential and induced mitochondrial fragmentation. Pre-treatment of cells with antioxidant, α-lipoic acid, normalised the ROS production and restored the mitochondrial membrane potential. These changes were accompanied with morphological changes of epithelial mesenchymal transition. CML-BSA increased the protein level of hypoxia-inducible factor-1α (HIF-1α), and the EMT-associated transcription factor, TWIST. These effects were reversed by α-lipoic acid. CML-BSA increased the protein levels of mesenchymal-specific markers, including vimentin, α-smooth muscle actin, which were alleviated by pre-treatment with α-lipoic acid. Our data suggest that CML-BSA induces EMT through a ROS/HIF-1α/TWIST-dependent mechanism, and that α-lipoic acid may alleviate the CML-induced EMT in renal tubular cells.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Lisina/análogos & derivados , Ácido Tióctico/farmacología , Células Cultivadas , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Túbulos Renales Proximales/metabolismo , Lisina/antagonistas & inhibidores , Lisina/farmacología , Especies Reactivas de Oxígeno/metabolismo
18.
Nutrients ; 10(12)2018 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-30513810

RESUMEN

Advanced glycation endproducts (AGEs) can be found in protein- and sugar-rich food products processed at high temperatures, which make up a vast amount of the Western diet. The effect of AGE-rich food products on human health is not yet clear and controversy still exists due to possible contamination of samples with endotoxin and the use of endogenous formed AGEs. AGEs occur in food products, both as protein-bound and individual molecules. Which form exactly induces a pro-inflammatory effect is also unknown. In this study, we exposed human macrophage-like cells to dietary AGEs, both in a protein matrix and individual AGEs. It was ensured that all samples did not contain endotoxin concentrations > 0.06 EU/mL. The dietary AGEs induced TNF-alpha secretion of human macrophage-like cells. This effect was decreased by the addition of N(ε)-carboxymethyllysine (CML)-antibodies or a receptor for advanced glycation endproducts (RAGE) antagonist. None of the individual AGEs induce any TNF-alpha, indicating that AGEs should be bound to proteins to exert an inflammatory reaction. These findings show that dietary AGEs directly stimulate the inflammatory response of human innate immune cells and help us define the risk of regular consumption of AGE-rich food products on human health.


Asunto(s)
Dieta Occidental/efectos adversos , Productos Finales de Glicación Avanzada/efectos adversos , Inflamación/inducido químicamente , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Anticuerpos/farmacología , Caseínas/química , Caseínas/farmacología , Endotoxinas/análisis , Contaminación de Alimentos/análisis , Productos Finales de Glicación Avanzada/farmacología , Glicosilación , Calor , Humanos , Lisina/análogos & derivados , Lisina/antagonistas & inhibidores , Lisina/inmunología , Macrófagos/inmunología , Reacción de Maillard , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Clin Invest ; 50(10): 2084-90, 1971 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-5165598

RESUMEN

Plasma thromboplastin antecedent (PTA, factor XI) is an important intermediate in the intrinsic coagulation system, and plasma kallikrein has been implicated as a mediator of the inflammatory process. Whereas their biologic activities are functionally distinct, their identity as separate entities in plasma has not been fully established, and the nature of their plasma inhibitors has not been completely characterized. A partially purified preparation containing the clotting, tosyl arginine methyl ester (TAMe) esterase and kinin-producing activities of these substances has been prepared by DEAE-cellulose chromatography of a Celite eluate obtained from acid-treated human plasma. These activities were not separable by acrylamide gel electrophoresis nor by isoelectric focusing, their pI being approximately 8.7. Human plasma alpha(2)-macroglobulin has been shown to inhibit the proteolytic activity of kallikrein and to inhibit partially its TAMe esterase activity. An alpha(2)-macroglobulin, PTA, kallikrein incubation mixture was separated by gel filtration chromatography. The alpha(2)-macroglobulin formed a high molecular weight complex with kallikrein and appeared in early chromatographic fractions. The PTA-clotting activity was not inhibited by the alpha(2)-macroglobulin; 64% of the initial PTA activity was isolated in later fractions free of kallikrein-induced kinin-like activity. In contrast, clotting, TAMe esterase, and kinin-forming activities were inhibited after gel filtration chromatography of an incubation mixture of these activities and partially purified C1 inactivator (C1 esterase inhibitor). Electrofocusing of an incubation mixture of an activated PTA, kallikrein preparation, and alpha(2)-macroglobulin resulted in the isolation of a PTA fraction free of kallikrein proteolytic activity, and with 4% of the original TAMe esterase activity. In this manner, activated PTA and plasma kallikrein have been shown to be distinct substances, and methods have been introduced for the further purification of active coagulation factor XI.


Asunto(s)
Factor XI/aislamiento & purificación , Indicadores y Reactivos , Calicreínas/antagonistas & inhibidores , Macroglobulinas , Animales , Arginina/antagonistas & inhibidores , Cromatografía DEAE-Celulosa , Cromatografía en Gel , Electroforesis Discontinua , Esterasas/antagonistas & inhibidores , Factor XI/análisis , Humanos , Focalización Isoeléctrica , Calicreínas/sangre , Lisina/antagonistas & inhibidores , Métodos , Ratas
20.
Diabetes Res Clin Pract ; 77(2): 337-40, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17383766

RESUMEN

Aspirin showed an inhibitory effect on the formation of pentosidine, a cross-linking advanced glycation endproduct, in collagen incubated with glucose in vitro. IC(50) was evaluated at 10mmol/l. Aspirin might act by metallic ion chelating (as did EDTA and DTPA) and by oxygen radical scavenging. Since aspirin was reported to inhibit retinopathy in diabetic dogs, it could act partly by inhibiting advanced glycation endproduct accumulation in long-lived proteins like collagens.


Asunto(s)
Arginina/análogos & derivados , Aspirina/farmacología , Colágeno/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Lisina/análogos & derivados , Arginina/antagonistas & inhibidores , Colágeno/efectos de los fármacos , Reactivos de Enlaces Cruzados , Glucosa/farmacología , Glicosilación/efectos de los fármacos , Lisina/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA