Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biochem Mol Toxicol ; 38(1): e23533, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37718616

RESUMEN

Humans are constantly exposed to low concentrations of ubiquitous environmental pollutant, Bisphenol A (BPA). Due to the prevalence of hypertension (one of the major risk factors of cardiovascular disease [CVD]) in the population, it is necessary to explore the adverse effect of BPA under hypertension associated pathogenic milieu. The current study exposed the Nω-nitro-l-arginine methyl ester (L-NAME) induced hypertensive Wistar rats to low dose BPA (50 µg/kg) for 30 days period. In tissue samples immunohistochemistry, real-time quantitative polymerase chain reaction and enzymatic assays were conducted. Moreover, studies on primary kidney cell culture were employed to explore the impact of low dose of BPA exposure at nanomolar level (20-80 nM range) on renal cells through various fluorescence assays. The observed results illustrate that BPA exposure potentiates/aggravates hypertension induced tissue abnormalities (renal fibrosis), oxidative stress (ROS generation), elevated angiotensin-converting enzyme activity, malfunction of the antioxidant and tricarboxylic acid cycle enzymes, tissue lipid abnormalities and inflammatory factor expression (both messenger RNA and protein level of TNF-α and IL-6). Further, in vitro exposure of nM levels of BPA to primary kidney cells modulates oxidative stress (both superoxide and total ROS), mitochondrial physiology (reduced mitochondrial transmembrane potential-∆ψm) and lipid peroxidation in a dose dependent manner. In addition, angiotensin II induced ROS generation was aggravated further by BPA during coexposure in kidney cells. Therefore, during risk assessment, a precise investigation on BPA exposure in hypertensive (CVD vulnerable) populations is highly suggested.


Asunto(s)
Angiotensina II , Hipertensión , Fenoles , Ratas , Humanos , Animales , Angiotensina II/farmacología , Especies Reactivas de Oxígeno/metabolismo , Ratas Wistar , Estrés Oxidativo , Riñón , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Compuestos de Bencidrilo/toxicidad
2.
Food Chem Toxicol ; 183: 114197, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38029875

RESUMEN

Human exposure to the hazardous chemical, Bisphenol A (BPA), is almost ubiquitous. Due to the prevalence of hypertension (CVD risk factor) in the aged human population, it is necessary to explore its adverse effect in hypertensive subjects. The current study exposed the Nω-nitro-l-arginine methyl ester (L-NAME) induced hypertensive Wistar rats to human exposure relevant low dose of BPA (50 µg/kg) for 30 days period. The liver biochemical parameters, histopathology, immunohistochemistry, gene expression (RT-qPCR), trace elements (ICP-MS), primary rat hepatocytes cell culture and metabolomic (1H NMR) assessments were performed. Results illustrate that BPA exposure potentiates/aggravates hypertension induced tissue abnormalities (hepatic fibrosis), oxidative stress, ACE activity, malfunction of the antioxidant system, lipid abnormalities and inflammatory factor (TNF-α and IL-6) expression. Also, in cells, BPA increased ROS generation, mitochondrial dysfunction and lipid peroxidation without any impact on cytotoxicity and caspase 3 and 9 activation. Notably, BPA exposure modulate lipid metabolism (cholesterol and fatty acid) in primary hepatocytes. Finally, the influence of ERK1/2, p38MAPK, ER stress and oxidative stress during relatively high dose of BPA elicited cytotoxicity was observed. Therefore, a precise hazardous risk investigation of BPA exposure in hypertensive populations is highly recommended.


Asunto(s)
Hipertensión , Hígado , Humanos , Ratas , Animales , Anciano , Ratas Wistar , Hepatocitos , Estrés Oxidativo , Compuestos de Bencidrilo/farmacología , Hipertensión/inducido químicamente
3.
Int J Biol Macromol ; 251: 126309, 2023 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-37573902

RESUMEN

In the present study, bacterial nanocellulose/graphene oxide nano-biocomposites (BNC-GO-NBCs) were fabricated by Komagataeibacter saccharivorans NUWB1 using an in-situ method involving three time-dependent approaches. Physicochemical studies showed that the chosen dried BNC-GO-NBC possessed a three-dimensional interconnected porous structure of BNC with GO layers embedded within the BNC fibrils. BNC-GO-NBC had a crystallinity index of 74.21 %, higher thermostability up to 380 °C and could withstand a tensile load of 84.72 MPa. N2 adsorption-desorption isotherm of the BNC-GO-NBC was found to be of type IV, suggesting a mesoporous type structure with a total pore volume and surface area of 6.232e-04 cc g-1 and 10.498 m2. BNC-GO-NBC exhibited remarkable adsorption capacity for two cationic dyes, Rhodamine B (RhB) and Acridine Orange (AO), and the adsorption data conformed well to the Langmuir isotherm (R2 = 0.99) and pseudo-second-order model. Thermodynamic studies indicated that the adsorption process was spontaneous and endothermic. Additionally, the BNC-GO-NBC displayed the potential for regeneration, with the ability to be recycled up to five times. Further, the antibacterial activity, cell cytotoxicity and oxidative stress assays of the BNC-GO-NBC revealed its non-cytotoxic nature. The findings of the present investigation evidently suggest the potentiality of BNC-GO-NBC in the application of dye adsorption and other environmental applications.

4.
Toxicology ; 485: 153414, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36587891

RESUMEN

Human exposure to plastic contaminated foods and environmental micro/nano plastic derived chemicals necessitates system-wide health risk assessment. Hence, current study intend to explore the mode of action (MoA) based adverse outcome pathways of 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), the major active metabolite of bisphenol A (BPA). The computational study employed broad range of target prediction, systems biology tools and molecular docking protocols. Further, validation of MBP targets was done using protein-ligand fluorescence quenching assay, endothelial cell culture and chicken embryo vascular angiogenesis models. Interestingly, the current results illustrate that various physiological signaling pathways (MAPK and VEGF related angiogenesis signaling) and disease progression pathways (hypertension, cancer and endocrine disorders) were enriched as potential targets of MBP. Further, docking studies highlights the possible binding mechanism of MBP with important targets including endothelial nitric oxide synthase (eNOS) and serum albumin (BSA). In addition, the validation studies on MBP-BSA interaction (fluorescence quenching), eNOS derived nitric oxide (NOx) generation in endothelial cells and chicken embryo angiogenesis support the system-wide impacts of MBP with highlights on cardiovascular pathogenesis. Thus, the current observation provides novel insights into the system wide impacts of MBP for the futuristic health risk assessment of plastic derived chemicals.


Asunto(s)
Contaminantes Ambientales , Animales , Humanos , Embrión de Pollo , Simulación del Acoplamiento Molecular , Células Endoteliales/metabolismo , Compuestos de Bencidrilo/química
5.
Environ Sci Pollut Res Int ; 29(36): 54250-54263, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35301628

RESUMEN

Recent evidences illustrated that the release of aluminum oxide nanoparticles (Al2O3-NPs) into the biosphere may pose risk to the environment and cause adverse effects on living organisms including humans. The current study assessed the hepatotoxic effects of Al2O3-NPs on developing chicken embryo and cell culture models. Results demonstrated that Al2O3-NPs exposure causes histological abnormalities and increased the level of tissue damage markers (ALP, AST, and ALT) in the embryonic liver. Furthermore, increased oxidative stress (TBARS) and impaired function of antioxidant enzymes (SOD, CAT, and GPx) were also observed. Moreover, it adversely affects red blood cells (RBC) morphology, liver metabolism, and stress response gene expression (HO-1 and NQO-1). Dose-dependent ROS generation and cytotoxic response in addition to potentiating effect on tumor necrosis factor alpha (TNF-α)-induced apoptosis (caspase-3 activity) were also observed. Inhibition of p38 mitogen-activated protein kinase (p38 MAPK) and c-Jun N-terminal kinase (JNK) pathways modulates Al2O3-NPs-induced apoptosis in HepG2 cells. Novel mechanisms behind embryonic hepatotoxicity, cytotoxic potentiating effects, and possible prevention strategies have been explored.


Asunto(s)
Óxido de Aluminio , Nanopartículas , Proteínas Quinasas p38 Activadas por Mitógenos , Óxido de Aluminio/toxicidad , Animales , Apoptosis , Embrión de Pollo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Nanopartículas/toxicidad , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
J Toxicol Environ Health A ; 85(6): 217-229, 2022 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-34706627

RESUMEN

The effects of environmental chemicals on health outcomes may be underestimated due to deficiency of knowledge regarding the actions of compounds on toxico-pathogenic mechanisms underlying biological systems outcomes. In this regard, the current study aimed to explore the potential target-pathway-disease relationship attributed to bisphenol A (BPA) responses in target tissues. Computational methods including reverse pharmacophore mapping approach, structural similarity based search and kinome wide interaction profiling were employed with molecular docking validation. Gene ontology (GO) enrichment analysis and protein-protein interaction (PPI) network based illustrations were utilized to prioritize target-pathway and disease relationships. Data illustrated that BPA possessed multi-target nature since this chemical potentially interacted with various protein targets where many of these were validated through docking. Potential BPA targets were significantly enriched to various cellular signaling pathways including steroid biosynthesis, peroxisome proliferator-activated receptor gamma (PPARℽ) and cancer. Further, hypertension was prioritized as disease target. In addition, BPA targeted 17 cell signaling kinases encompassed in the human kinome. In addition, inflammatory (5-LO) and apoptosis regulators (Bcl-X and Bcl-2) were also explored as novel targets. Evidence indicates that the multi-target nature and plausible mechanisms underlying BPA actions in a system wide manner aids toward understanding of adverse effects. This observation may lead us to more precise method to elucidate the toxico-pathogenic mechanisms of BPA with an environmental health perspective.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Disruptores Endocrinos/toxicidad , Fenoles/toxicidad , Transducción de Señal , Compuestos de Bencidrilo/química , Contaminantes Ambientales/toxicidad , Ontología de Genes , Humanos , Simulación del Acoplamiento Molecular , Fenoles/química , Proteoma
7.
Chem Biol Interact ; 351: 109719, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34699767

RESUMEN

The precise toxico-pathogenic effects of zinc oxide nanoparticles (ZnO-NPs) on the cardiovascular system under normal and cardiovascular disease (CVD) risk factor milieu are unclear. In this study, we have investigated the dose-dependent effects of ZnO-NPs on developing chicken embryo and cell culture (H9c2 cardiomyoblast, HUVEC and aortic VSMC) models. In addition, the potentiation effect of ZnO-NPs on simulated risk factor conditions was evaluated using; 1. Reactive oxygen species (ROS) induced cardiac remodeling, 2. Angiotensin-II induced cardiac hypertrophy, 3. TNF-α induced HUVEC cell death and 4. Inorganic phosphate (Pi) induced aortic VSMC calcification models. The observed results illustrates that ZnO-NPs exposure down regulates vascular development and elevates oxidative stress in heart tissue. At the cellular level, ZnO-NPs exposure reduced the cell viability and increased the intracellular ROS generation, lipid peroxidation and caspase-3 activity in a dose-dependent manner in all three cell types. In addition, ZnO-NPs exposure significantly suppressed the endothelial nitric oxide (NO) generation, cardiac Ca2+ - ATPase activity and enhanced the cardiac mitochondrial swelling. Moreover, inhibition of p38 MAPK and JNK signaling pathways influence the cytotoxicity. Overall, ZnO-NPs exposure affects the cardiovascular system under normal conditions and it exacerbates the cardiovascular pathogenesis under selected risk factor milieu.


Asunto(s)
Cardiomegalia/metabolismo , Cardiotoxinas/toxicidad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Estrés Oxidativo/efectos de los fármacos , Óxido de Zinc/toxicidad , Animales , Apoptosis/efectos de los fármacos , Cardiomegalia/inducido químicamente , Cardiotoxicidad , Pollos , Embrión no Mamífero/efectos de los fármacos , Corazón/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mitocondrias/efectos de los fármacos , Ratas
8.
Hum Exp Toxicol ; 40(12_suppl): S654-S665, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34797181

RESUMEN

Due to the prevalence of hypertension (one of the major risk factors of CVD) in the population, it is necessary to explore the adverse effects of daily tolerable and "safe" dose of bisphenol A (BPA) under hypertensive conditions. The current study exposed the Nω-nitro-l-arginine methyl ester (L-NAME, 40 mg/kg b.w/day) induced hypertensive Wistar rats to BPA (50 µg/kg b.w/day) by oral administration along with appropriate controls for 30 days period. The results illustrate that a 'safe' dose of BPA does not influence the systolic blood pressure (SBP) and levels of circulatory biomarkers of tissue damage. On the other hand, BPA exposure significantly (p < 0.05) elevates the thiobarbituric acid reactive substances (TBARS) content in plasma and tissues (heart, aorta, liver and kidney) in hypertensive rats when compared with respective control (BPA alone exposed) rats. Similarly, a significant modulation of ROS generation in RBC, plasma nitric oxide (NO) level and angiotensin-converting enzyme (ACE) activity was observed only under hypertensive milieu. In conclusion, the observed adverse effects during 'safe' dose of BPA exposure are specific to the hypertensive condition. Therefore, a precise investigation to explore the effects of BPA exposure in vulnerable hypertensive populations is highly suggested.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Hipertensión/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fenoles/toxicidad , Animales , Presión Sanguínea/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
9.
Gene Rep ; 25: 101312, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34401607

RESUMEN

Coronavirus disease 2019 (COVID-19) is a viral pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that led to more than 800,00 deaths and continues to be a major threat worldwide. The scientific community has been studying the risk factors associated with SARS-CoV-2 infection and pathogenesis. Recent studies highlight the possible contribution of atmospheric air pollution, specifically particulate matter (PM) exposure as a co-factor in COVID-19 severity. Hence, meaningful translation of suitable omics datasets of SARS-CoV-2 infection and PM exposure is warranted to understand the possible involvement of airborne exposome on COVID-19 outcome. Publicly available transcriptomic data (microarray and RNA-Seq) related to COVID-19 lung biopsy, SARS-CoV-2 infection in epithelial cells and PM exposure (lung tissue, epithelial and endothelial cells) were obtained in addition with proteome and interactome datasets. System-wide pathway/network analysis was done through appropriate software tools and data resources. The primary findings are; 1. There is no robust difference in the expression of SARS-CoV-2 entry factors upon particulate exposure, 2. The upstream pathways associated with upregulated genes during SARS-CoV-2 infection considerably overlap with that of PM exposure, 3. Similar pathways were differentially expressed during SARS-CoV-2 infection and PM exposure, 4. SARS-CoV-2 interacting host factors were predicted to be associated with the molecular impact of PM exposure and 5. Differentially expressed pathways during PM exposure may increase COVID-19 severity. Based on the observed molecular mechanisms (direct and indirect effects) the current study suggests that airborne PM exposure has to be considered as an additional co-factor in the outcome of COVID-19.

10.
Chem Res Toxicol ; 32(4): 589-602, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30834740

RESUMEN

Thalidomide is an infamous teratogen and it is continuously being explored for its anticancer properties. Fibroblast growth factor receptors (FGFRs) are implicated in embryo development and cancer pathophysiology. With striking similarities observed between FGFR implicated conditions and thalidomide embryopathy, we hypothesized thalidomide targets FGFRs. We utilized three different cell lines and chicken embryo model to investigate the effects of thalidomide and analogs on FGFR expression. We performed molecular docking, KINOMEscan analysis, and kinase activity assays to study the drug-protein interactions. The expression of FGFR1 and FGFR2 was differentially regulated by all the three drugs in cells as well as in developing organs. Transcriptome analysis of thalidomide-treated chick embryo strongly suggests the modulation of FGFR signaling and key transcription factors. Corroboration with previous studies suggests that thalidomide might affect FGFR expression through the transcription factor, E2F1. At the protein level, molecular docking predicted all three analogs to interact with lysine residue at 517th and 508th positions of FGFR2 and FGFR3, respectively. This lysine coordinates the ATP binding site of FGFR, thus hinting at the possible perturbation of FGFR activity by thalidomide. Kinome analysis revealed that kinase activities of FGFR2 and FGFR3 (G697C) reduced by 31% and 65%, respectively, in the presence of 10 µM thalidomide. Further, we checked and confirmed that the analogs inhibited the FGFR2 kinase activity in a dose-dependent manner. This study suggests that FGFRs could be potential targets of thalidomide and the two analogs, and also endorses the link between the teratogenicity and antitumor activities of the drugs.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Talidomida/análogos & derivados , Talidomida/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Pollos , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Relación Estructura-Actividad , Talidomida/química
11.
RSC Adv ; 8(36): 20211-20221, 2018 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-35541641

RESUMEN

Onco-cardiology is critical for the management of cancer therapeutics since many of the anti-cancer agents are associated with cardiotoxicity. Therefore, the major aim of the current study is to employ a novel in silico method combined with experimental validation to explore off-targets and prioritize the enriched molecular pathways related to the specific cardiovascular events other than their intended targets by deriving relationship between drug-target-pathways and cardiovascular complications in order to help onco-cardiologists for the management of strategies to minimize cardiotoxicity. A systems biological understanding of the multi-target effects of a drug requires prior knowledge of proteome-wide binding profiles. In order to achieve the above, we have utilized PharmMapper, a web-based tool that uses a reverse pharmacophore mapping approach (spatial arrangement of features essential for a molecule to interact with a specific target receptor), along with KEGG for exploring the pathway relationship. In the validation part of the study, predicted protein targets and signalling pathways were strengthened with existing datasets of DrugBank and antibody arrays specific to vascular endothelial growth factor (VEGF) signalling in the case of 5-fluorouracil as direct experimental evidence. The current systems toxicological method illustrates the potential of the above big-data in supporting the knowledge of onco-cardiological indications which may lead to the generation of a decision making catalogue in future therapeutic prescription.

12.
Chem Res Toxicol ; 30(10): 1883-1896, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-28892372

RESUMEN

Since the conception of thalidomide as a teratogen, approximately 30 hypotheses have been put forward to explain the developmental toxicity of the molecule. However, no systems biology approach has been taken to understand the phenomena yet. The proposed work was aimed to explore the mechanism of thalidomide toxicity in developing chick embryo in the context of transcriptomics by using genome wide RNA sequencing data. In this study, we challenged the developing embryo at the stage of blood island formations (HH8), which is the most vulnerable stage for thalidomide-induced deformities. We observed that thalidomide affected the early vasculogenesis through interfering with the blood island formation extending the effect to organogenesis. The transcriptome analyses of the embryos collected on sixth day of incubation showed that liver, eye, and blood tissue associated genes were down regulated due to thalidomide treatment. The conserved gene coexpression module also indicated that the genes involved in lens development were heavily affected. Further, the Gene Ontology analysis explored that the pathways of eye development, retinol metabolism, and cartilage development were dampened, consistent with the observed deformities of various organs. The study concludes that thalidomide exerts its toxic teratogenic effects through interfering with early extra-embryonic vasculogenesis and ultimately gives an erroneous transcriptomic pattern to organogenesis.


Asunto(s)
Perfilación de la Expresión Génica , Neovascularización Patológica/genética , Organogénesis/genética , Talidomida/toxicidad , Animales , Embrión de Pollo , Neovascularización Patológica/inducido químicamente , Organogénesis/efectos de los fármacos , Talidomida/administración & dosificación
13.
J Pharm Biomed Anal ; 141: 250-261, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28463780

RESUMEN

Omega-3 fatty acids are clinically useful and the two marine omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are prevalent in fish and fish oils. Omega-3 fatty acid formulations should undergo a rigorous regulatory step in order to obtain United States Food and Drug Administration (USFDA) approval as prescription drug. In connection with that, despite quantifying EPA and DHA fatty acids, there is a need for quantifying the level of ethyl esters of them in biological samples. In this study, we make use of reverse phase high performance liquid chromatography coupled with mass spectrometry (RP-HPLC-MS)technique for the method development. Here, we have developed a novel multiple reaction monitoring method along with optimized parameters for quantification of EPA and DHA as ethyl esters. Additionally, we attempted to validate the bio-analytical method by conducting the sensitivity, selectivity, precision accuracy batch, carryover test and matrix stability experiments. Furthermore, we also implemented our validated method for evaluation of pharmacokinetics of omega fatty acid ethyl ester formulations.


Asunto(s)
Espectrometría de Masas en Tándem , Cromatografía Liquida , Ácidos Docosahexaenoicos , Ácido Eicosapentaenoico , Ésteres , Ácidos Grasos Omega-3
14.
Mol Biosyst ; 12(12): 3683-3694, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27752677

RESUMEN

Since there is no precise therapy for treating vascular calcification by directly targeting the vascular wall, we aim to unveil novel drug targets through mining the molecular effect of a high phosphate environment on vascular cells through computational methods. Here, we hypothesize that manipulation of the vascular pathogenic network by small molecule therapeutics predicted from prior knowledge might offer great promise. With this, we intend to understand the publicly available transcriptomic data of vascular smooth muscle cells and endothelial cells exposed to the high phosphate induced vascular calcification milieu and to re-examine the above published experiments for reasons different from those examined in the previous studies through multilevel systems biological understanding. Hence, in this study the differentially expressed genes were subjected to both upstream and downstream network analysis through multiple standalone software and web servers. To provide an insight into causal signaling, we simultaneously predicted upstream regulatory layers through transcription factor and kinome enrichment analysis. Moreover the possible systems pharmacological choices were presented in three ways as (1) drug induced expression modulation, (2) drugs that interact with upstream and downstream regulatory targets, (3) possible natural product therapeutics from target-compound relationship. Furthermore for validating the current study we have specifically evaluated the preventive effect of two predicted natural compounds in a bovine aortic calcification model. The overall observation predicts a few novel mechanisms that might be involved in vascular dysfunction and calcification in both cell types. Also, the systems pharmacological investigation provides clues for the possible therapeutic options along with validation. In conclusion, the current study indicates that reanalysis of transcriptomic data propels us to reposition the approved drugs and use natural compounds as novel therapeutic agents for vascular calcification.


Asunto(s)
Redes Reguladoras de Genes , Transducción de Señal , Biología de Sistemas/métodos , Calcificación Vascular/etiología , Calcificación Vascular/metabolismo , Animales , Bovinos , Análisis por Conglomerados , Biología Computacional , Descubrimiento de Drogas , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Transcriptoma , Calcificación Vascular/tratamiento farmacológico
15.
Sci Rep ; 6: 27304, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27255968

RESUMEN

Disturbed fluid flow or modulated shear stress is associated with vascular conditions such as atherosclerosis, thrombosis, and aneurysm. In vitro simulation of the fluid flow around the plaque micro-environment remains a challenging approach. Currently available models have limitations such as complications in protocols, high cost, incompetence of co-culture and not being suitable for massive expression studies. Hence, the present study aimed to develop a simple, versatile model based on Computational Fluid Dynamics (CFD) simulation. Current observations of CFD have shown the regions of modulated shear stress by the disturbed fluid flow. To execute and validate the model in real sense, cell morphology, cytoskeletal arrangement, cell death, reactive oxygen species (ROS) profile, nitric oxide production and disturbed flow markers under the above condition were assessed. Endothelium at disturbed flow region which had been exposed to low shear stress and swirling flow pattern showed morphological and expression similarities with the pathological disturbed flow environment reported previously. Altogether, the proposed model can serve as a platform to simulate the real time micro-environment of disturbed flow associated with eccentric plaque shapes and the possibilities of studying its downstream events.


Asunto(s)
Endotelio Vascular/fisiología , Placa Aterosclerótica/fisiopatología , Estrés Mecánico , Simulación por Computador
16.
Eur J Pharmacol ; 777: 113-23, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26945821

RESUMEN

The present study was designed to evaluate the effect of sinapic acid, a bioactive phenolic acid on high blood pressure associated cardiac dysfunction, kidney fibrosis and lipid alterations in N(ω)-nitro-l-arginine methyl ester hydrochloride (l-NAME) induced hypertensive rats. Sinapic acid was administered to rats orally at a dosage of 40 mg/kg everyday for a period of 4 weeks. Sinapic acid treatment significantly decreased mean arterial pressure, left ventricular end diastolic pressure, organ weights (liver and kidney), lipid peroxidation products in tissues (liver and kidney), activities of hepatic marker enzymes and the levels of renal function markers in serum of l-NAME rats. Sinapic acid treatment also significantly increased the level of plasma nitric oxide metabolites, and enzymatic and non-enzymatic antioxidants in tissues of l-NAME rats. Tissue damage was assessed by histopathological examination. Alterations in plasma angiotensin-converting enzyme activity, level of plasma lipoproteins and tissue lipids were corrected by sinapic acid treatment in l-NAME rats. Sinapic acid treatment significantly decreased the activity of 3-hydroxy-3-methylglutaryl-Coenzyme A (HMG-CoA) reductase in plasma and liver, whereas the activity of lecithin cholesterol acyl transferase was significantly increased in the plasma of hypertensive rats. Docking result showed the interaction between sinapic acid and HMG-CoA reductase. Sinapic acid has shown best ligand binding energy of -5.5 kcal/M. Moreover, in chick embryo model, sinapic acid improved vessel density on chorioallantoic membrane. These results of the present study concludes that sinapic acid acts as a protective agent against hypertension associated cardiac dysfunction, kidney fibrosis and lipid alterations.


Asunto(s)
Acilcoenzima A/metabolismo , Ácidos Cumáricos/farmacología , Corazón/efectos de los fármacos , Corazón/fisiopatología , Hipertensión/metabolismo , Riñón/patología , NG-Nitroarginina Metil Éster/farmacología , Acilcoenzima A/química , Animales , Antioxidantes/metabolismo , Presión Sanguínea/efectos de los fármacos , Ácidos Cumáricos/metabolismo , Fibrosis , Hipertensión/fisiopatología , Riñón/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Lipoproteínas/sangre , Lipoproteínas/metabolismo , Masculino , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/enzimología , Mitocondrias Hepáticas/metabolismo , Simulación del Acoplamiento Molecular , Óxido Nítrico/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Conformación Proteica , Ratas , Ratas Wistar
17.
Br J Nutr ; 114(9): 1385-94, 2015 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-26346559

RESUMEN

In our previous studies, veratric acid (VA) shows beneficial effect on hypertension and its associated dyslipidaemia. In continuation, this study was designed to investigate the effect of VA, one of the major benzoic acid derivatives from vegetables and fruits, on cardiovascular remodelling in hypertensive rats, primarily assessed by functional studies using Langendorff isolated heart system and organ bath system. Hypertension was induced in male albino Wistar rats by oral administration of N ω -nitro-l-arginine methyl ester hydrochloride (l-NAME) (40 mg/kg body weight (b.w.)) in drinking water for 4 weeks. VA was orally administered at a dose of 40 mg/kg b.w. l-NAME-treated rats showed impaired cardiac ventricular and vascular function, evaluated by Langendorff isolated heart system and organ bath studies, respectively; a significant increase in the lipid peroxidation products such as thiobarbituric acid-reactive substances and lipid hydroperoxides in aorta; and a significant decrease in the activities of superoxide dismutase, catalase, glutathione peroxidase and levels of GSH, vitamin C and vitamin E in aorta. Fibrotic remodelling of the aorta and heart were assessed by Masson's Trichrome staining and Van Gieson's staining, respectively. In addition, l-NAME rats showed increased heart fibronectin expression assessed by immunohistochemical analysis. VA supplementation throughout the experimental period significantly normalised cardiovascular function, oxidative stress, antioxidant status and fibrotic remodelling of tissues. These results of the present study conclude that VA acts as a protective agent against hypertension-associated cardiovascular remodelling.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Frutas/química , Hipertensión/tratamiento farmacológico , Ácido Vanílico/análogos & derivados , Remodelación Vascular/efectos de los fármacos , Verduras/química , Administración Oral , Animales , Antioxidantes/administración & dosificación , Aorta/efectos de los fármacos , Aorta/metabolismo , Ácido Ascórbico/metabolismo , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/metabolismo , Catalasa/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Peróxidos Lipídicos/metabolismo , Masculino , NG-Nitroarginina Metil Éster/administración & dosificación , NG-Nitroarginina Metil Éster/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Ácido Vanílico/administración & dosificación , Vitamina E/metabolismo
18.
Biochem Biophys Res Commun ; 456(4): 853-9, 2015 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-25511706

RESUMEN

The present study was designed to evaluate antioxidant and cardioprotective potential of sinapic acid (SA) against ischemia/reperfusion (I/R) injury. Cardiac functional recovery after I/R was evaluated by percentage rate pressure product (%RPP) and percentage coronary flow (%CF). Myocardial injury was evaluated by 2,3,5-triphenyltetrazolium chloride (TTC) staining and LDH enzyme leakage. Oxidative stress was estimated by lipid peroxidation level. eNOS protein expression in reperfused heart was assessed using Western blot method. Finally, in order to support the antioxidant effect of SA, in vitro protective potential of SA was assessed on H2O2-induced oxidative stress in H9c2 cardiomyoblast cells. The overall results demonstrated that I/R induced cardiac dysfunction, injury and oxidative stress was attenuated by SA treatment. Moreover, in vitro results also shown that, SA protects H9c2 cells from oxidative stress and modulates mitochondrial membrane permeability transition (MPT). In conclusion, coupled results from both in vivo and in vitro experiments have confirmed that SA with antioxidant role protects cardiac cells and its functions from I/R induced oxidative stress.


Asunto(s)
Cardiotónicos/uso terapéutico , Ácidos Cumáricos/uso terapéutico , Mioblastos/patología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/patología , Estrés Oxidativo , Animales , Cardiotónicos/farmacología , Ácidos Cumáricos/farmacología , Peróxido de Hidrógeno , Técnicas In Vitro , L-Lactato Deshidrogenasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/enzimología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Recuperación de la Función/efectos de los fármacos
19.
PLoS One ; 9(12): e115682, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25531679

RESUMEN

OBJECTIVES: Hypertensive heart disease is a constellation of abnormalities that includes cardiac fibrosis in response to elevated blood pressure, systolic and diastolic dysfunction. The present study was undertaken to examine the effect of sinapic acid on high blood pressure and cardiovascular remodeling. METHODS: An experimental hypertensive animal model was induced by L-NAME intake on rats. Sinapic acid (SA) was orally administered at a dose of 10, 20 and 40 mg/kg body weight (b.w.). Blood pressure was measured by tail cuff plethysmography system. Cardiac and vascular function was evaluated by Langendorff isolated heart system and organ bath studies, respectively. Fibrotic remodeling of heart and aorta was assessed by histopathologic analyses. Oxidative stress was measured by biochemical assays. mRNA and protein expressions were assessed by RT-qPCR and western blot, respectively. In order to confirm the protective role of SA on endothelial cells through its antioxidant property, we have utilized the in vitro model of H2O2-induced oxidative stress in EA.hy926 endothelial cells. RESULTS: Rats with hypertension showed elevated blood pressure, declined myocardial performance associated with myocardial hypertrophy and fibrosis, diminished vascular response, nitric oxide (NO) metabolites level, elevated markers of oxidative stress (TBARS, LOOH), ACE activity, depleted antioxidant system (SOD, CAT, GPx, reduced GSH), aberrant expression of TGF-ß, ß-MHC, eNOS mRNAs and eNOS protein. Remarkably, SA attenuated high blood pressure, myocardial, vascular dysfunction, cardiac fibrosis, oxidative stress and ACE activity. Level of NO metabolites, antioxidant system, and altered gene expression were also repaired by SA treatment. Results of in vitro study showed that, SA protects endothelial cells from oxidative stress and enhance the production of NO in a concentration dependent manner. CONCLUSIONS: Taken together, these results suggest that SA may have beneficial role in the treatment of hypertensive heart disease by attenuating fibrosis and oxidative stress through its antioxidant potential.


Asunto(s)
Antiinfecciosos/uso terapéutico , Presión Sanguínea/efectos de los fármacos , Enfermedades Cardiovasculares/prevención & control , Ácidos Cumáricos/uso terapéutico , Hipertensión/prevención & control , Óxido Nítrico/metabolismo , Remodelación Ventricular/efectos de los fármacos , Animales , Antioxidantes/metabolismo , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/fisiopatología , Células Cultivadas , Inhibidores Enzimáticos/toxicidad , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Masculino , NG-Nitroarginina Metil Éster/toxicidad , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/genética , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
20.
Biochimie ; 102: 183-7, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24742379

RESUMEN

Cardiovascular dysfunction and vascular calcification is the leading cause of death in chronic renal failure (CRF) patients. This study was designed to evaluate the effect of diosgenin on coronary flow resistance and to address the question whether the previously proven antivascular calcification potential of diosgenin is associated or not with the osteochondrogenic transdifferentiation of vascular smooth muscle cells (VSMC). In this study, CRF in Wistar rats was induced by fed with 0.75% adenine and diosgenin was treated everyday at the dose of 40 mg/kg. Langendorff based isolated heart protocol was employed to analyze the coronary flow resistance. Western blot method was used to explore the phosphorylation dynamics of endothelial nitric oxide synthase (eNOS) at the serine 1177 residue. In addition, cardiac nitric oxide metabolites level also assessed. Quantitative expression of VSMC and osteochondrogenic markers was also evaluated. Antioxidant potential of diosgenin was studied in vitro. The outcome of the present study explores that diosgenin treatment significantly improves the coronary resistance and increased the nitric oxide metabolites level compared with CRF. Further, diosgenin increases the phosphorylation of eNOS (peNOS ser1177). Moreover, diosgenin reduced the aortic expression of osteochondrogenic markers and improved the VSMC phenotype components. Further, diosgenin shows concentration dependent antioxidant potential. In conclusion, this study have proven that diosgenin have enough potential to improve the coronary function and interfere the osteochondrogenic transdifferentiation program of aortic VSMC which supports its antivascular calcification potential.


Asunto(s)
Calcinosis/tratamiento farmacológico , Diosgenina/administración & dosificación , Fallo Renal Crónico/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/administración & dosificación , Antioxidantes/metabolismo , Aorta/citología , Aorta/efectos de los fármacos , Calcinosis/metabolismo , Calcinosis/patología , Transdiferenciación Celular/genética , Vasos Coronarios/citología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/patología , Endotelio Vascular/efectos de los fármacos , Humanos , Fallo Renal Crónico/patología , Músculo Liso Vascular/citología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...