Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
ACS Omega ; 9(2): 2783-2794, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38250361

RESUMEN

The emergence of multidrug-resistant (MDR) bacteria has spurred the exploration of therapeutic nanomaterials such as ZnO nanoparticles. However, the inherent nonspecific toxicity of ZnO has posed a significant obstacle to their clinical utilization. In this research, we propose a novel approach to improve the selectivity of the toxicity of ZnO nanoparticles by impregnating them onto a less toxic clay mineral, Bentonite, resulting in ZB nanocomposites (ZB NCs). We hypothesize that these ZB NCs not only reduce toxicity toward both normal and carcinogenic cell lines but also retain the antibacterial properties of pure ZnO nanoparticles. To test this hypothesis, we synthesized ZB NCs by using a precipitation technique and confirmed their structural characteristics through X-ray diffraction and Raman spectroscopy. Electron microscopy revealed composite particles in the size range of 20-50 nm. The BET surface area of ZB NCs, within a relative pressure (P/P0) range of 0.407-0.985, was estimated to be 31.182 m2/g. Notably, 50 mg/mL ZB NCs demonstrated biocompatibility with HCT 116 and HEK 293 cell lines, supported by flow cytometry and fluorescence microscopy analysis. In vitro experiments further confirmed a remarkable five-log reduction in the population of MDR Escherichia coli in the presence of 50 mg/mL of ZB NCs. Antibacterial activity of the nanocomposites was also validated in the HEK293 and HCT 116 cell lines. These findings substantiate our hypothesis and underscore the effectiveness of ZB NCs against MDR E. coli while minimizing nonspecific toxicity toward healthy cells.

3.
Biotechnol Genet Eng Rev ; : 1-32, 2023 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-37226482

RESUMEN

One of the most essential chemical processes that is utilized in the manufacturing of a great deal of contemporary goods is called heterogeneously catalyzed reactions, and it is also one of the most fascinating. Metallic nanostructures are heterogeneous catalysts for range reactions due to their huge surface area, large assembly of active surface sites, and quantum confinement effects. Unprotected metal nanoparticles suffer from irreversible agglomeration, catalyst poisoning, and limited life cycle. To circumvent these technical disadvantages, catalysts are frequently spread on chemically inert materials like as mesoporous Al2O3, ZrO2, and different types of ceramic material. In this research, plentiful bauxite residue is used to create a low-cost alternative catalytic material. We have hydrogenated p-Nitrophenol to p-Aminophenol on bauxite residue (BR) supported silver nanocomposites (Ag NCs). The phase and crystal structure, bond structure and morphological analysis of the developed material will be done XRD, FTIR, and SEM-EDX respectively. The ideal conditions were 150 ppm of catalyst, 0.1 mM of p-NP, and 10 minutes overall up-to 99% conversion of p-NP to p-AP. A multi-variable predictive model created using Response Surface Methodology (RSM) and a data-based Artificial Neural Network (ANN) model were found to be the best ways to predict the maximum conversion efficiency. ANN models predicted efficiency more accurately than RSM models, and the strong agreement between model predictions and experimental data was indicated by their low relative error (RE0.10), high regression coefficient (R2>0.97), and Willmott-d index (dwill-index > 0.95) values.

4.
Biosci Rep ; 42(11)2022 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-36149310

RESUMEN

Medicinal plants (MPs) are natural sources of active compounds with potential therapeutic benefits in alleviating various illnesses for decades. Fijian people also are using these MPs for the management/prevention of Type 2 diabetes mellitus (T2DM) and associated complications. However, till date, none of these Fijian MP's antidiabetic potential have been explored or evaluated. Here, we investigated the antidiabetic potential of Fijian MPs scientifically. Phytochemicals such as polyphenols were detected to inhibit the activity of α-amylase and α-glucosidase, the two key carbohydrate enzymes linked to T2DM. Therefore, in the present study, the total phenolic content (TPC), α-amylase and α-glucosidase inhibitory activity of five Fijian MPs: Vobo (Mussaenda raiateensis, MR), Vula walu (Blechnum orientale, BO), Gasau (Miscanthus floridulus, MF), Molikaro (Citrus limon, CL) and Beki ni sina (Dicranopteris caudate, DC) collected from mainland region of Vitilevu, Fiji Islands, were evaluated in vitro. The hydromethanolic (ME) and dichloromethane (DM) extracts of these selected MPs were investigated. The ME extracts of BO (0.102 ± 0.009 mM CE) and DC (0.098 ± 0.09 mM Catechin Equivalence [CE]) showed a higher TPC compared with the control [vanillic acid (0.052 ± 0.003 mM CE, *P value < 0.05)]. However, the TPC of MF, MR and CL were found in the range of 0.020 ± 0.009 to 0.009 ± 0.01 mM CE. The ME extracts of MF and MR inhibited α-glucosidase significantly in comparison with acarbose as evidenced from the IC50 values (IC50 of MF = 1.58 ± 0.03 ng/µl; IC50 of MR = 1.87 ± 0.43 ng/µl and IC50 of acarbose = 3.34 ± 0.15 ng/µl). Moreover, DM extracts of MR (IC50 = 1.31 ± 0.29 ng/µl) also showed significantly higher α-glucosidase inhibitory activity. In contrary, MR (IC50 = 16.18 ± 0.16 ng/µl) and CL (IC50 = 9.21 ± 0.51 ng/µl) also showed significant α-amylase inhibitory activity in ME and DM extracts, respectively. These, results suggest that Fijian MPs could be a potential source of natural inhibitors of enzymes involved in carbohydrate digestion and thus may possibly be used in managing T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2 , Plantas Medicinales , Plantas Medicinales/química , Diabetes Mellitus Tipo 2/tratamiento farmacológico , alfa-Glucosidasas/uso terapéutico , Inhibidores de Glicósido Hidrolasas/farmacología , Inhibidores de Glicósido Hidrolasas/química , Inhibidores de Glicósido Hidrolasas/uso terapéutico , Acarbosa/uso terapéutico , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Extractos Vegetales/química , alfa-Amilasas , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Hipoglucemiantes/química , Fenoles/farmacología
5.
PLoS One ; 16(12): e0260835, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34882718

RESUMEN

Myocardial infarction (MI), atherosclerosis and other inflammatory and ischemic cardiovascular diseases (CVDs) have a very high mortality rate and limited therapeutic options. Although the diagnosis is based on markers such as cardiac Troponin-T (cTrop-T), the mechanism of cTrop-T upregulation and release is relatively obscure. In the present study, we have investigated the mechanism of cTrop-T release during acute hypoxia (AH) in a mice model by ELISA & immunohistochemistry. Our study showed that AH exposure significantly induces the expression and release of sterile inflammatory as well as MI markers in a time-dependent manner. We further demonstrated that activation of TLR3 (mediated by eRNA) by AH exposure in mice induced cTrop-T release and Poly I:C (TLR3 agonist) also induced cTrop-T release, but the pre-treatment of TLR3 immuno-neutralizing antibody or silencing of Tlr3 gene or RNaseA treatment two hrs before AH exposure, significantly abrogated AH-induced Caspase 3 activity as well as cTrop-T release. Our immunohistochemistry and Masson Trichrome (MT) staining studies further established the progression of myocardial injury by collagen accumulation, endothelial cell and leukocyte activation and adhesion in myocardial tissue which was abrogated significantly by pre-treatment of RNaseA 2 hrs before AH exposure. These data indicate that AH induced cTrop-T release is mediated via the eRNA-TLR3-Caspase 3 pathway.


Asunto(s)
Apoptosis , Vesículas Extracelulares/genética , Hipoxia/fisiopatología , Infarto del Miocardio/patología , ARN/genética , Receptor Toll-Like 3/metabolismo , Troponina T/metabolismo , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Ratones , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , ARN/metabolismo , Transducción de Señal , Receptor Toll-Like 3/genética
6.
J Inflamm Res ; 13: 823-828, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33149655

RESUMEN

Coronavirus disease 2019 (COVID-19) has spread rapidly throughout the world. The range of the disease is broad but among hospitalized patients with COVID-19 are coagulation disorders, pneumonia, respiratory failure, and acute respiratory distress syndrome (ARDS). The excess production of early response proinflammatory cytokines results in what has been described as a cytokine storm, leading to an increased risk of thrombosis, inflammations, vascular hyperpermeability, multi-organ failure, and eventually death over time. As the pandemic is spreading and the whole picture is not yet clear, we highlight the importance of coagulation disorders in COVID-19 infected subjects and summarize it. COVID-19 infection could induce coagulation disorders leading to clot formation as well as pulmonary embolism with detrimental effects in patient recovery and survival. Coagulation and inflammation are closely related. In this review, we try to establish an association between virus infections associated with innate immune activation, inflammation and coagulation activation.

7.
Blood Cells Mol Dis ; 84: 102459, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32559654

RESUMEN

Sterile Inflammation (SI), a condition where damage associated molecular patterns (DAMPs) released from dying cells, leads to TLR (Toll-like receptor) activation and triggers hypoxemia in circulation leading to venous thrombosis (VT) through tissue factor (TF) activation, but its importance under acute hypoxia (AH) remains unexplored. Thus, we hypothesized that eRNA released from dying cells under AH activates TF via the TLR3-ERK1/2-AP1 pathway, leading to VT. Animals were exposed to stimulate hypoxia for 0-24 h at standard temperature and humidity. RNaseA and DNase1 were injected immediately before exposure. TLR3 gene silencing was performed through in vivo injection of TLR3 siRNA. 80 µg/kg BW of isolated eRNA and eDNA were injected 6 h prior to sacrifice. Antigens of TF pathway were determined by ELISA and TF activity by a chromogenic assay. AH exposure significantly induced release of SI markers i.e. eRNA, eDNA, HMGB1 and upregulated TLR3, ERK1/2 (Extracellular signal-regulated kinases), AP1 (Activator Protein-1) and TF, whereas RNaseA pre-treatment diminished the effect of AH, thus inhibiting TF expression as well as activity during AH. Hence, we propose a possible mechanism of AH-induced TF activation and thrombosis where RNaseA can become the novel focal point in ameliorating therapy for AH induced thrombosis.


Asunto(s)
Hipoxia/metabolismo , ARN/metabolismo , Proteína de Replicación C/metabolismo , Transducción de Señal , Tromboplastina/metabolismo , Receptor Toll-Like 3/metabolismo , Animales , Células Cultivadas , Hipoxia/complicaciones , Hipoxia/genética , Sistema de Señalización de MAP Quinasas , Ratones , Tromboplastina/genética , Trombosis/etiología , Trombosis/genética , Trombosis/metabolismo , Regulación hacia Arriba
8.
Sci Rep ; 9(1): 11175, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31371738

RESUMEN

Sterile inflammation (SI) is an essential process in response to snakebite and injury. The venom induced pathophysiological response to sterile inflammation results into many harmful and deleterious effects that ultimately leads to death. The available treatment for snakebite is antiserum which does not provide enough protection against venom-induced pathophysiological changes like haemorrhage, necrosis, nephrotoxicity and often develop hypersensitive reactions. In order to overcome these hindrances, scientists around the globe are searching for an alternative therapy to provide better treatment to the snake envenomation patients. In the present study TiO2 (Titanium dioxide)-NPs (Nanoparticles) has been assessed for antisnake venom activity and its potential to be used as an antidote. In this study, the synthesis of TiO2-NPs arrays has been demonstrated on p-type Silicon Si < 100 > substrate (∼30 ohm-cm) and the surface topography has been detected by Field-emission scanning electron microscopy (FESEM). The TiO2-NPs successfully neutralized the Daboia russelii venom (DRV) and Naja kaouthia venom (NKV)-induced lethal activity. Viper venom induced haemorrhagic, coagulant and anticoagulant activities were effectively neutralized both in in-vitro and in vivo studies. The cobra and viper venoms-induced sterile inflammatory molecules (IL-6, HMGB1, HSP70, HSP90, S100B and vWF) were effectively neutralised by the TiO2-NPs in experimental animals.


Asunto(s)
Inflamación/prevención & control , Nanopartículas/uso terapéutico , Fosfolipasas A2/metabolismo , Venenos de Serpiente/antagonistas & inhibidores , Titanio/uso terapéutico , Animales , Animales de Laboratorio , Venenos Elapídicos , Hemorragia/prevención & control , Ratones , Nanopartículas/química , Necrosis/prevención & control , Mordeduras de Serpientes/patología , Mordeduras de Serpientes/fisiopatología , Mordeduras de Serpientes/terapia , Venenos de Serpiente/toxicidad , Venenos de Víboras
9.
J Leukoc Biol ; 105(4): 705-717, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30835912

RESUMEN

PGE2 plays a critical role in angiogenesis, ischemic, and neuro-inflammatory disorders of the brain, which breakdown the blood-brain barrier (BBB). However, the effects of PGE2 on human brain endothelial cell (HBECs) migration, a key process in the angiogenic response and BBB stability, are not well defined. In this study, we investigated the mechanism of PGE2 in HBECs migration in vitro. Here we showed that PGE2 stimulated migration of HBECs in a dose-time and matrix-dependent manner, evaluated by the Boyden chamber assay, but other prostanoids failed to do so. PGE2 receptor (EP2; butaprost), EP3 (sulprostone), and EP4 (PGE1 -OH) receptor agonists stimulated HBECs migration, but the silencing of EP significantly attenuated this effect. EP1 agonist (11-trinor PGE1 ) had no effect on HBECs migration on silencing of the EP1 receptor. We further showed that PGE2 stimulated cAMP production and activated protein kinase A (PKA), whereas pretreatment with the adenyl cyclase inhibitor (dideoxyadenosine; 1 µM) or PKA inhibitors, H89 (0.5 µM)/PKAI (1 µM), completely abrogated PGE2 -induced migration. Furthermore, silencing of the EP2/EP4 receptors significantly inhibited PGE2 -induced cAMP and PKA activation, whereas EP3 receptor silencing failed to do so. These results suggest that PGE2 regulates HBEC migration via cooperation of EP2, EP3, and EP4 receptors. Coupling of PGE2 to these receptors resulted in increased production of cAMP, which regulates HBEC migration via PKA pathway. The elucidation of molecular events involved is critical for the development of targeted strategies to treat cerebrovascular diseases associated with dysregulated angiogenesis.


Asunto(s)
Encéfalo/citología , Movimiento Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dinoprostona/farmacología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Receptores de Prostaglandina E/metabolismo , Movimiento Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , AMP Cíclico/biosíntesis , Células Endoteliales/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos
10.
Biomed Pharmacother ; 111: 1302-1314, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30841444

RESUMEN

INTRODUCTION: Garlic has been reported to stimulate nitric-oxide (NO) synthesis in various cells. The role of aqueous-extract of garlic (AEG) and a purified NO-generating protein from garlic (NGPG) was investigated to control hyperglycemia by hepatic insulin synthesis through NGPG induced synthesis of NO via glucose-activated NO-synthase and glucose transporter-4 (Glut-4) in the hepatocytes. METHODS: Type-1-diabetic mellitus mice were prepared by alloxan treatment, NO was determined by methemoglobin method, insulin synthesis was quantitated by ELISA. TNF-α and NFκß was quantitated by ELISA. The AEG-induced Glut-4 synthesis was determined by in-vitro translation of mRNA from the hepatocytes. The NO-generating protein from AEG was purified to homogeneity by chromatography on DEAE-cellulose and Sephadex G-50 columns and sequenced/characterized by Mass-spectral-analysis. RESULTS: Purified NGPG injection to diabetic mice significantly reduced the blood-sugar and increase insulin level in diabetic animal. It also increases insulin-release, Glut-4 synthesis, glucose-uptake in both liver and ß-cells of diabetic mice. NGPG down regulated pro-inflammatory cytokine TNF-α and the stress responsive NFκB-expression in liver cell of diabetic mice. Injection of AEG to the diabetic mice reduced the blood glucose level from 550 ± 10 mg/dL to 125 ± 10 mg/dL in 16 h with simultaneous increase of plasma NO from 0 nmol/h to 2.5 nmol/h and insulin 2 ± 1.1µunit/mL to 15µunit/mL at 16 h. Oral administration of AEG to adult diabetic mice increased NO, insulin and Glut-4 synthesis in the hepatocytes. CONCLUSION: AEG and the purified-NGPG protein can control hyperglycemia through the stimulation of NO by glucose-activated NO-synthase that would play an important role in the synthesis of insulin/Glut-4 in liver-cells.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Ajo/química , Transportador de Glucosa de Tipo 4/metabolismo , Hipoglucemiantes/farmacología , Insulina/metabolismo , Hígado/efectos de los fármacos , Óxido Nítrico/metabolismo , Aloxano/farmacología , Animales , Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Glucosa/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Hígado/metabolismo , Ratones , Extractos Vegetales/farmacología
11.
J Vasc Res ; 55(4): 244-254, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30223272

RESUMEN

High mobility group box 1 protein (HMGB1), a sterile inflammatory molecule and damage-associated molecular pattern (DAMP) released from various cells during stress has been implicated in inflammation. Several reports show that there is a direct relationship between inflammation and cardiovascular diseases (CVDs) such as thrombosis, hypertension, insulin resistance, preeclampsia, etc. Here, we intend to summarize the concept of the emerging link between HMGB1 and CVDs. Furthermore, we will discuss the possible therapeutic strategies that target HMGB1 for the treatment of different CVDs.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Proteína HMGB1/fisiología , Inflamación/fisiopatología , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedad Coronaria/fisiopatología , Coagulación Intravascular Diseminada/fisiopatología , Femenino , Proteína HMGB1/antagonistas & inhibidores , Humanos , Oxidación-Reducción , Preeclampsia/fisiopatología , Embarazo , Transducción de Señal , Accidente Cerebrovascular/fisiopatología , Trombosis de la Vena/fisiopatología
12.
Curr Mol Med ; 18(10): 672-678, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30734678

RESUMEN

BACKGROUND: An increase in the level of cytokines like TNF-α and IL-6 causes the inflammatory surge in acute ischemic heart disease (AIHD). OBJECTIVE: A high-level dermcidin isoform-2 (DCN-2) occurrence in AIHD was subjected to determine a possible regulation of cytokines expression. The effect of estrogen to counteract the inflammatory response was determined. METHODS: Blood was collected from AIHD patients and normal volunteers with consent. Nitric oxide (NO) synthesis was done with methemoglobin method.TNF-α and IL-6 expression were determined by ELISA and Western blot. RESULTS: (DCN-2) incubation with 120nM to the normal neutrophil solution for 2h resulted in the increase of TNF-α from 3.82±1.53pg/ml to 20.7±6.9pg/ml and IL-6 from 3.27±1.52pg/ml to 47.07±3.4pg/ml. In AIHD patients, the cytokine level was18.3- 27.3pg/ml, with a median value 21.86pg/ml (TNF-α) and IL-6 level was 23.54- 52.73pg/ml, with a median value 42.16pg/ml. Treatment with 0.6nM estriol, a kind of female steroid hormone estrogen for 45min decreased the elevated cytokine level in 120nM DCN-2 treated normal neutrophils. DCN-2 induced TNF-α synthesis in neutrophils was further determined by Western blot technique with a thickened band intensity of TNF-α. Estriol (0.6nM) treatment also influenced the DCN-2 induced inhibition of nitric oxide (NO) synthesis from 0nmol NO/ml to 0.56nmol/ml. The subsequent reduction of TNF-α level correlates the increase of NO level. CONCLUSION: In conclusion, the stress-induced DCN-2 production in AIHD propagates the inflammatory response. Steroid molecule like estriol plays a protective role by reducing DCN-2 responses in the NO synthesis.


Asunto(s)
Estriol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-6/biosíntesis , Isquemia Miocárdica/metabolismo , Neutrófilos/metabolismo , Óxido Nítrico/biosíntesis , Péptidos/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Adulto , Femenino , Humanos , Masculino , Isquemia Miocárdica/patología , Neutrófilos/patología , Isoformas de Proteínas/biosíntesis
13.
Eur J Immunol ; 46(10): 2388-2400, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27480067

RESUMEN

Increased plasma level of von Willebrand Factor (vWF) is associated with major cardiovascular diseases. We previously reported that multimeric vWF binds to NO synthase and inhibits insulin-induced production of NO, thus promoting insulin resistance during acute hypoxia (AH). However, the transcriptional regulation of vWF during AH is not clearly understood. Here, we investigated the mechanisms underlying the upregulation of vwf in mice. AH significantly upregulates the tlr2, tlr3, myd88, and vwf expression and phosphorylation of specificity protein 1 (SP1). Furthermore, AH significantly upregulates high mobility group box-1 (HMGB1) in a time-dependent manner. Moreover, a TLR2 agonist upregulates vWF but a TLR3 agonist does not. Pretreatment with an HMGB1 inhibitor, TLR2-immunoneutralizing antibody, or SP1 inhibitor significantly inhibits vWF expression. Furthermore, Tlr2 silencing completely inhibited MYD88, vWF expression, and SP1 phosphorylation. However, pretreatment with glycyrrhizic acid or silencing of Tlr2 completely blocks binding of Sp1 to the Vwf promoter, thus inhibiting its expression, and enhances insulin resistance during AH. Patients with type 2 diabetes mellitus also showed significantly elevated levels of HMGB1, TLR2, SP1, and vWF, thereby supporting the results of the murine model of AH. Taken together, HMGB1 upregulates vWF in vivo through the TLR2-MYD88-SP1 pathway in mice.


Asunto(s)
Diabetes Mellitus Tipo 2/inmunología , Proteína HMGB1/metabolismo , Hipoxia/inmunología , Factor de von Willebrand/metabolismo , Animales , Anticuerpos Bloqueadores/administración & dosificación , Células Cultivadas , Diabetes Mellitus Tipo 2/terapia , Ácido Glicirrínico/farmacología , Proteína HMGB1/genética , Humanos , Hipoxia/terapia , Inmunoglobulinas/metabolismo , Resistencia a la Insulina , Ratones , Terapia Molecular Dirigida , Factor 88 de Diferenciación Mieloide/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Regulación hacia Arriba
14.
Sci Rep ; 6: 26789, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27226415

RESUMEN

A trypsin resistant oral insulin preparation was made by incubating insulin for 2 h at 23 °C with previously boiled cow milk at 100 °C that was coagulated with 0.6 M acetic acid. The precipitate was resuspended in the same volume of milk. The immunoblot analysis of the suspended proteins treated with 200 ng of trypsin/ml for 3 h demonstrated that the 80.1% of the insulin in the suspension survived the proteolytic degradation compared to 0% of the hormone survived in the control. The feeding of 0.4 ml (0.08 unit of insulin) of the resuspended proteins followed by 0.2 ml of the same protein to alloxan induced diabetic mice maximally decreased the blood glucose level from 508 ± 10 mg/dl to 130 ± 10 mg/dl in 7 h with simultaneous increase of the basal plasma concentration of insulin from 3 ± 1.1 µunits/ml to 18 ± 1.5 µunits/ml. In control experiment the absence of insulin in the identical milk suspension produced no hypoglycemic effect suggesting milk was not responsible for the hypoglycemic effect of milk-insulin complex. Coming out of insulin-casein complex from the intestinal gut to the circulation was spontaneous and facilitated diffusion transportation which was found from Gibbs free energy reaction.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Insulina/uso terapéutico , Administración Oral , Aloxano , Animales , Glucemia/análisis , Caseínas/metabolismo , Bovinos , Estabilidad de Medicamentos , Insulina/sangre , Insulina/química , Insulina/farmacocinética , Intestino Delgado/metabolismo , Ratones , Leche , Unión Proteica , Distribución Tisular , Tripsina/metabolismo
15.
Eur J Immunol ; 45(11): 3158-73, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26350442

RESUMEN

Endogenous ligands released from dying cells, including extracellular RNA (eRNA), cause TLR activation, which is associated with inflammation and vascular diseases. However, the importance of this response in acute hypoxia (AH) remains unexplored. Here, we observed eRNA-mediated TLR3 activation during exposure of mice to AH in the absence of exogenous viral stimuli. RNaseA treatment diminished AH-induced expression of IFN and cell adhesion molecules (CAMs) and myeloid cell infiltration in the lung, and TLR3 gene silencing or neutralization with antibodies markedly attenuated AH- or poly I:C-induced IFN and CAM expression and leukocyte adhesion (LA) and myeloid cell infiltration in the lung. However, RNaseA treatment or TLR3 gene silencing failed to alter AH-induced cell death and proliferation in lung vasculature. Furthermore, IFN-γ--but not IFN-α--regulated AH-induced CAM expression and LA. Treatment with RNaseA, TLR3 siRNA, neutralizing antibodies, or a STAT1 inhibitor substantially decreased AH- and poly I:C-induced STAT1 phosphorylation, CAM expression, and myeloid cell infiltration, suggesting a central role for STAT1 phosphorylation in AH-induced LA and infiltration. We conclude that eRNA activates TLR3 and facilitates, through in vivo IFN-γ-STAT1 signaling, AH-induced leukocyte infiltration in the lung. Thus, RNaseA might provide a therapeutic alternative for patients with lung diseases.


Asunto(s)
Hipoxia/patología , Leucocitos/metabolismo , Pulmón/patología , ARN/metabolismo , Transducción de Señal/inmunología , Animales , Western Blotting , Quimiotaxis de Leucocito/fisiología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Hipoxia/inmunología , Inmunohistoquímica , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interferón gamma/metabolismo , Pulmón/inmunología , Ratones , ARN/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/metabolismo , Receptor Toll-Like 3/metabolismo
16.
Biochemistry ; 53(1): 115-26, 2014 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-24328140

RESUMEN

Hypoxic respiratory diseases or hypoxia exposures are frequently accompanied by glucose intolerance and impaired nitric oxide (NO) availability. However, the molecular mechanism responsible for impaired NO production and insulin resistance (IR) during hypoxia remains obscure. In this study, we investigated the possible mechanism of impaired NO production and IR during hypoxia in a mouse model. Mice were exposed to hypoxia for different periods of time (0-24 h), and parameters of IR and endothelial dysfunctions were analyzed. Exposure to hypoxia resulted in a time-dependent increase in IR as well as multimeric forms of von Willebrand factor (vWF) and subsequently a decrease in eNOS activity. Preincubation with plasma of hypoxia-exposed animals (different time points) or human vWF inhibited insulin-induced NO production in a dose-dependent manner; larger doses of insulin reversed the effect. In contrast, preincubation of vWF-immunodepleted plasma failed to inhibit insulin-induced NO production, whereas vWF immunoneutralization abolished the effect of hypoxia-induced IR and D-[U-(14)C]glucose uptake. Furthermore, the interactions between vWF and eNOS were studied by far-Western blotting, co-immunoprecipitation, and surface plasma resonance spectroscopy. Kinetic analyses showed that the dissociation constant (KD), inhibitory constant (Ki), and half-maximal inhibitory concentration (IC50) were 1.79 × 10(-8) M, 250 pM, and 18.31 pM, respectively, suggesting that vWF binds to eNOS with a high affinity and greater efficacy for activator (insulin) inhibition. These results indicated that vWF, an antagonist of eNOS, inhibits insulin-induced NO production and causes IR.


Asunto(s)
Hipoxia/fisiopatología , Resistencia a la Insulina/fisiología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Factor de von Willebrand/fisiología , Animales , Eritrocitos/metabolismo , Glucosa/metabolismo , Humanos , Concentración 50 Inhibidora , Insulina/farmacología , Cinética , Ratones
17.
Blood Cells Mol Dis ; 49(2): 92-101, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22647506

RESUMEN

Hypoxemia in the circulation can lead to venous thrombosis (VT) through tissue factor (TF) activation, but the mechanism of TF activation in hypoxia remains obscure. Ligands released from damaged tissues or cells due to hypoxia are identified by various pattern-recognition receptors (PRR), including Toll-like receptor3 (TLR3). In the present study, we investigated the mechanism of TF activation during acute hypoxia in a rat model. The expression of TLR3 and TF was analyzed by immunoblotting and RT-PCR. The TF activity was evaluated by two-stage chromogenic assay and fibrin deposition was detected by immunohistochemistry. The expression of TLR3, TF, and TF activity was increased significantly 6 h post acute hypoxia and then decreased gradually. The contribution of TLR3 in TF activation was investigated by poly I:C and TLR3 neutralizing antibody. We also found increased ERK phosphorylation both in acute hypoxia and poly I:C treatment. We further showed that the pre-treatment of TLR3 neutralizing antibody or ERK inhibitor (PD98059) 2 h prior to acute hypoxia or poly I:C treatment completely abrogated ERK phosphorylation and TF activation. The pre-treatment of TLR3 neutralizing antibody also inhibited fibrin deposition in lung vasculature. These data indicate that acute hypoxia induced TF activation is mediated through TLR3-ERK1/2 pathway.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipoxia/metabolismo , Tromboplastina/agonistas , Receptor Toll-Like 3/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/genética , Fibrina/metabolismo , Flavonoides/farmacología , Regulación de la Expresión Génica , Hipoxia/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Fosforilación/efectos de los fármacos , Poli I-C/farmacología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Tromboplastina/genética , Tromboplastina/metabolismo , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 3/genética
18.
J Thromb Thrombolysis ; 33(3): 218-29, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22238031

RESUMEN

Insulin inhibits platelet aggregation through nitric oxide synthesis by stimulating platelet insulin activated nitric oxide synthase. Impaired platelet insulin activated nitric oxide synthase in acute myocardial infarction (AMI) patients had been reported and thus our aim was to identify and isolate the factors impairing insulin activated nitric oxide in acute myocardial infarction patients' plasma and study its effect on platelets aggregation in vitro. The insulin activated nitric oxide synthase inhibitor was identified as a protein and was purified from the plasma of AMI subjects using DEAE cellulose and Sephadex G-50 column, molecular weight determined by SDS-PAGE, nitric oxide quantified by methaemoglobin method, inhibitor protein quantified in plasma by immunoblot and ELISA, platelet aggregation studies done using an aggregometer, thromboxane-A2 in the platelets determined by radioimmunoassay, (125)I-insulin radioligand binding studies done using normal subject platelets. The purified nitric oxide synthase inhibitor protein was ~66 kDa, concentration in AMI subjects' plasma varied from 114 to 9,090 µM and was undetected in normal subjects' plasma. The inhibitor protein competes with insulin for insulin receptor binding sites. The Incubation of the normal subject PRP with 5.0 µM inhibitor for 30 min followed by 0.4 µM ADP addition caused platelet aggregation in vitro, 130 µM aspirin or 400 µU insulin/ml addition was able to abrogate 0.4 µM ADP induced platelet aggregation even in the presence of 5.0 µM inhibitor. A potent inhibitory protein against insulin activated nitric oxide synthase in platelets appears in circulation of AMI subjects impairing nitric oxide production, potentiating ADP induced platelet aggregation and increasing the thromboxane-A2 level in platelets.


Asunto(s)
Proteínas Sanguíneas/aislamiento & purificación , Proteínas Sanguíneas/metabolismo , Insulina/metabolismo , Infarto del Miocardio/sangre , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Adulto , Anciano , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Insulina/farmacología , Masculino , Persona de Mediana Edad , Infarto del Miocardio/metabolismo , Unión Proteica/fisiología
19.
J Environ Pathol Toxicol Oncol ; 31(4): 357-65, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23394448

RESUMEN

The development of mitigating agents to counter injuries induced by radiation is as important as the development of radioprotective agents. This study reports the ability of diallyl sulphide (DAS), a naturally occurring organosulfur compound, to mitigate radiation-induced mortality and injuries to the hematopoietic system in whole-body irradiated mice. Intraperitoneal administration of a single dose of DAS (160 mg/kg body weight) 2 hours after 9 Gy whole-body irradiation resulted in 37% animal survival as opposed to 100% mortality in the irradiation-only group, improved general conditions with no visual signs of sickness, and body weight loss. Restoration of spleen body weight index by DAS in animals exposed to sublethal dosages (2 and 5 Gy) of whole-body irradiation increased endogenous spleen colony-forming units and increased bone marrow cellularity indicated enhanced hematopoietic recovery, which was supported further by recoveries in lymphocyte count. Further studies to elucidate the mechanism of action of DAS are warranted to design effective protocols for mitigation of radiation injuries using compounds like DAS.


Asunto(s)
Compuestos Alílicos/farmacología , Hematopoyesis/efectos de la radiación , Protectores contra Radiación/farmacología , Sulfuros/farmacología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/efectos de la radiación , Hematopoyesis/efectos de los fármacos , Recuento de Leucocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Bazo/efectos de la radiación , Irradiación Corporal Total
20.
Mol Endocrinol ; 25(8): 1431-43, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21659473

RESUMEN

Extravillous trophoblasts (EVT) of the human placenta invade the uterine decidua and its arteries to ensure successful placentation. We previously identified two decidua-derived molecules, TGF-ß and a TGF-ß-binding proteoglycan decorin (DCN), as negative regulators of EVT proliferation, migration, and invasiveness and reported that DCN acts via multiple tyrosine kinase receptors [epidermal growth factor-receptor (EGF-R), IGF receptor-1 (IGFR1), and vascular endothelial growth factor 2 receptor (VEGFR-2)]. Because binding of DCN to VEGFR-2 has never been reported earlier, present study explored this binding, the approximate location of VEGFR-2-binding site in DCN, and its functional role in our human first trimester EVT cell line HTR-8/SVneo. Based on far-Western blotting and coimmunoprecipitation studies, we report that DCN binds both native (EVT expressed) and recombinant VEGFR-2 and that this binding is abrogated with a VEGFR-2 blocking antibody, indicating an overlap between the ligand-binding and the DCN-binding domains of VEGFR-2. We determined that (125)I-labeled VEGF-E (a VEGFR-2 specific ligand) binds EVT with a dissociation constant (K(d)) of 566 pM, and DCN displaced this binding with an inhibition constant (K(i)) of 3.93-5.78 nM, indicating a 7- to 10-fold lower affinity of DCN for VEGFR-2. DCN peptide fragments derived from the leucine rich repeat 5 domain that blocked DCN-VEGFR-2 interactions or VEGF-E binding in EVT cells also blocked VEGF-A- and VEGF-E-induced EVT cell proliferation and migration, indicative of functional VEGFR-2-binding sites of DCN. Finally, DCN inhibited VEGF-E-induced EVT migration by interfering with ERK1/2 activation. Our findings reveal a novel role of DCN as an antagonistic ligand for VEGFR-2, having implications for pathophysiology of preeclampsia, a trophoblast hypoinvasive disorder in pregnancy, and explain its antiangiogenic function.


Asunto(s)
Decorina/metabolismo , Trofoblastos/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Far-Western Blotting , Bovinos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Decorina/química , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Antígenos HLA-G/metabolismo , Humanos , Inmunoprecipitación , Ligandos , Modelos Biológicos , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Trofoblastos/enzimología , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...