Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Biochem Funct ; 42(2): e3942, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38379263

RESUMEN

Colorectal cancer (CRC) is among the most prevalent gastrointestinal cancers of epithelial origin worldwide, with over 2 million cases detected every year. Emerging evidence suggests a significant increase in the levels of inflammatory and stress-related markers in patients with CRC, indicating that oxidative stress and lipid peroxidation may influence signalling cascades involved in the progression of the disease. However, the precise molecular and cellular basis underlying CRC and their modulations during bioactive compound exposure have not yet been deciphered. This study examines the effect of caffeic acid phenethyl ester (CAPE), a natural bioactive compound, in HT29 CRC cells grown under serum-supplemented and serum-deprived conditions. We found that CAPE inhibited cell cycle progression in the G2/M phase and induced apoptosis. Migration assay confirmed that CAPE repressed cancer invasiveness. Protein localisation by immunofluorescence microscopy and protein expression by western blot analysis reveal increased expressions of key inflammatory signalling mediators such as p38α, Jun N-terminal kinase and extracellular signal-regulated kinase (ERK) proteins. Molecular docking data demonstrates that CAPE shows a higher docking score of -5.35 versus -4.59 to known p38 inhibitor SB203580 as well as a docking score of -4.17 versus -3.86 to known ERK1/2 inhibitor AZD0364. Co-immunoprecipitation data reveals that CAPE treatment effectively downregulates heat shock protein (HSP) expression in both sera-supplemented and limited conditions through its interaction with mitogen-activated protein kinase 14 (MAPK14). These results suggest that stress induction via serum starvation in HT29 CRC cells leads to the induction of apoptosis and co-ordinated activation of MAPK-HSP pathways. Molecular docking studies support that CAPE could serve as an effective inhibitor to target p38 and MAPK compared to their currently known inhibitors.


Asunto(s)
Neoplasias del Colon , Alcohol Feniletílico , Alcohol Feniletílico/análogos & derivados , Humanos , Línea Celular Tumoral , Proteínas de Choque Térmico , Simulación del Acoplamiento Molecular , Apoptosis , Ácidos Cafeicos/farmacología , Ácidos Cafeicos/metabolismo , Alcohol Feniletílico/farmacología , Alcohol Feniletílico/metabolismo , Neoplasias del Colon/tratamiento farmacológico
2.
Front Oncol ; 13: 1283431, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37746255
3.
World J Gastroenterol ; 29(16): 2433-2451, 2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37179581

RESUMEN

Ferroptosis is an emerging novel form of non-apoptotic, regulated cell death that is heavily dependent on iron and characterized by rupture in plasma membrane. Ferroptosis is distinct from other regulated cell death modalities at the biochemical, morphological, and molecular levels. The ferroptotic signature includes high membrane density, cytoplasmic swelling, condensed mitochondrial membrane, and outer mitochondrial rupture with associated features of accumulation of reactive oxygen species and lipid peroxidation. The selenoenzyme glutathione peroxidase 4, a key regulator of ferroptosis, greatly reduces the lipid overload and protects the cell membrane against oxidative damage. Ferroptosis exerts a momentous role in regulating cancer signaling pathways and serves as a therapeutic target in cancers. Dysregulated ferroptosis orchestrates gastrointestinal (GI) cancer signaling pathways leading to GI tumors such as colonic cancer, pancreatic cancer, and hepatocellular carcinoma. Crosstalk exists between ferroptosis and other cell death modalities. While apoptosis and autophagy play a detrimental role in tumor progression, depending upon the factors associated with tumor microenvironment, ferroptosis plays a decisive role in either promoting tumor growth or suppressing it. Several transcription factors, such as TP53, activating transcription factors 3 and 4, are involved in influencing ferroptosis. Importantly, several molecular mediators of ferroptosis, such as p53, nuclear factor erythroid 2-related factor 2/heme oxygenase-1, hypoxia inducible factor 1, and sirtuins, coordinate with ferroptosis in GI cancers. In this review, we elaborated on key molecular mechanisms of ferroptosis and the signaling pathways that connect ferroptosis to GI tumors.


Asunto(s)
Carcinoma Hepatocelular , Ferroptosis , Neoplasias Gastrointestinales , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patología , Peroxidación de Lípido , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Neoplasias Hepáticas/patología , Microambiente Tumoral
4.
Crit Rev Immunol ; 42(5): 21-41, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37075017

RESUMEN

Idiopathic pulmonary fibrosis (IPF) entails complex pathophysiological processes and complicated mechanisms. It is a type of lung disease that has no known cure. The disease's chronic inflammatory response is triggered by the abnormal activation of alveolar cells that create mediators that promote the development of myofibroblast and fibroblast foci. Usually, there is an excessive level of collagens and extracellular matrix deposition that lead to the destruction of the lung's architecture. The cause and pathogenesis of IPF are relatively complicated and unknown. The role of inflammation in the pathogenesis of IPF is still controversial. If only inflammation was the only crucial element to the disease events, lung fibrosis pathology would mean an influx of inflammatory cells, and the disease would act in response to immunosuppression. However, neither of these is true. Recent studies indicate that the pathophysiology of the disease is more a consequence of fibroblast dysfunction than poorly modulated inflammation. A broad range of factors has been recognized as crucial mediators in fibrosis. This article does not intend to deliver a comprehensive review of the molecular mechanisms in IPF but will concentrate on specific topics relating to IPF pathogenesis with relevance to immune modulation. In addition, we focus on the key mediators driving the pathogenesis of pulmonary fibrosis irrespective of their etiology, in conjunction with an overview of how these studies can be translated into appropriate or future diagnostic/therapeutic applications.


Asunto(s)
Fibrosis Pulmonar Idiopática , Humanos , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/patología , Inflamación/patología , Fibroblastos , Matriz Extracelular , Pulmón/patología
5.
Drug Chem Toxicol ; 45(3): 1264-1275, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-32924642

RESUMEN

Idiopathic Pulmonary Fibrosis (IPF) is a grave disease characterized by abnormal wound healing associated with chronic, progressive, irreversible fatal lung disease, leading to persistent injuries to the alveolar epithelium. A consequent disturbance of fibroblast proliferation and apoptosis results in subsequent release of pro-inflammatory and pro-fibrotic mediators coupled with accumulation of extracellular matrix within the interstitium. Inexorable distortion of lung alveolar architecture leads to respiratory failure with a median survival rate of 3-5 years. Currently available drugs can only slowdown the progression of fibrosis and novel drugs are warranted to treat this disease. In this study, we demonstrate the fibro-protective effect of diosgenin in experimental lung fibrosis through regulation of Epithelial Mesenchymal Transition (EMT). A single dose of 3 U/kg body weight (b.wt) Bleomycin (BLM) was administered intratracheally in Wistar male albino rats and fibrotic animals were treated with diosgenin (100 mg/kg b.wt) orally for 28 days. BLM administered rat show histological alteration with increased mast cell and collagen accumulation. BLM induced abnormalities were significantly reduced upon treatment with diosgenin. Western blot analysis revealed an increased level of pro-inflammatory and pro-fibrotic molecules such as IL-1ß and TGF-ß in BLM induced rats. Rats supplemented with diosgenin showed a decreased expression of inflammatory and pro-fibrotic mediators. Markers of EMT molecules were evaluated by immunoblot. The results of immunoblot demonstrate that diosgenin regulated the expression of TGF-ß mediated EMT. Hence, from the overall study, administration of diosgenin prevents pulmonary fibrosis by restraint inflammation and EMT.


Asunto(s)
Diosgenina , Fibrosis Pulmonar , Animales , Bleomicina/toxicidad , Diosgenina/metabolismo , Diosgenina/farmacología , Diosgenina/uso terapéutico , Transición Epitelial-Mesenquimal , Pulmón , Masculino , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/prevención & control , Ratas , Ratas Wistar , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta/uso terapéutico , Factor de Crecimiento Transformador beta1/metabolismo
6.
Life Sci ; 264: 118657, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33148421

RESUMEN

AIM: Indomethacin [IND] is reported to treat colon cancer. However, continuous exposure to IND causes gastric ulceration, an adverse side effect in humans. This study implies the therapeutic effect of IND and juglone [JUG] against colon carcinogenesis, without gastric ulceration - an adverse side effect of IND. MATERIALS AND METHODS: Adult male Balb/C mice were divided into six groups randomly: control, AOM/DSS-induced, IND-treated, JUG-treated, IND + JUG-treated and drug-control. Levels of serum markers, haematoxylin & eosin staining to observe tissue architecture, toluidine blue staining to detect mast cells expression, Masson's trichrome and sirius-red staining were used to detect the collagen deposition. RT-PCR and western blot analysis were carried out to detect inflammation and apoptosis. KEY FINDINGS: IND + JUG effectively decreased the levels of serum markers: CEA, AFP, LDH, AST and ALT. Although, IND restored colonic architecture by regulating the accumulation of mast cell and collagen content, it causes gastric ulceration. To address this adverse effect of IND, JUG was given along with IND and was shown to alleviate IND-induced gastric ulceration. AOM/DSS induced animals showed increased expression of inflammatory molecules - TNFα, NFκB and Cox-2, apoptosis regulator - Bcl-2 and decreased expression of pro-apoptotic molecules - Bad, Bax and caspase3; whereas, IND and JUG treated groups showed decreased inflammatory expression with increased expression of pro-apoptotic molecules. SIGNIFICANCE: IND and JUG reduce the inflammatory activity and induce apoptotic cell death, while JUG effectively prevents IND induced gastric ulceration. These findings establish that a combination of IND + JUG may serve as a promising treatment regimen for colon cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinogénesis/patología , Neoplasias del Colon/patología , Indometacina/farmacología , Inflamación/patología , Naftoquinonas/farmacología , Animales , Azoximetano , Carcinogénesis/efectos de los fármacos , Recuento de Células , Línea Celular Tumoral , Colágeno/metabolismo , Sulfato de Dextran , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/patología , Ratones Endogámicos BALB C
7.
J Biochem Mol Toxicol ; 34(2): e22433, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31916655

RESUMEN

Colorectal cancer (CRC) is the third most common fatal cancer. Indomethacin, a nonsteroidal anti-inflammatory drug, is known to reduce the occurrence of CRC. This study evaluated the potential anticolon cancer effects of juglone (5-hydroxy-1,4-naphthoquinone) in combination with indomethacin. Human colon adenocarcinoma cells (HT29) were subjected to treatment with indomethacin, juglone, and a combination of both. Morphological analysis, cell cycle regulation, and dual staining using acridine orange and ethidium bromide in control and treated cells revealed the apoptotic potential of these compounds. Bcl2 and inflammatory molecules (tumor necrosis factor-α, nuclear factor kappa B, and Cox-2) were found to be decreased with a concomitant increase in the expression of proapoptotic molecules (Bad, Bax, cytochrome c, and PUMA) as a result of the molecular regulation of Wnt, Notch, and peroxisome proliferator-activated receptor-γ signaling. Treatment with juglone was not as effective as with indomethacin; however, a combination of both was shown to be more effective, suggesting that juglone may be considered for therapeutic intervention of colon cancer.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/metabolismo , Indometacina/farmacología , Mediadores de Inflamación/metabolismo , Naftoquinonas/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Antineoplásicos/uso terapéutico , Proteínas Reguladoras de la Apoptosis/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Sinergismo Farmacológico , Células HT29 , Humanos , Indometacina/uso terapéutico , Concentración 50 Inhibidora , Naftoquinonas/uso terapéutico , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Vía de Señalización Wnt
8.
World J Gastrointest Oncol ; 10(9): 244-259, 2018 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-30254720

RESUMEN

Colorectal carcinogenesis (CRC) imposes a major health burden in developing countries. It is the third major cause of cancer deaths. Despite several treatment strategies, novel drugs are warranted to reduce the severity of this disease. Adenomatous polyps in the colon are the major culprits in CRC and found in 45% of cancers, especially in patients 60 years of age. Inflammatory polyps are currently gaining attention in CRC, and a growing body of evidence denotes the role of inflammation in CRC. Several experimental models are being employed to investigate CRC in animals, which include the APCmin/+ mouse model, Azoxymethane, Dimethyl hydrazine, and a combination of Dextran sodium sulphate and dimethyl hydrazine. During CRC progression, several signal transduction pathways are activated. Among the major signal transduction pathways are p53, Transforming growth factor beta, Wnt/ß-catenin, Delta Notch, Hippo signalling, nuclear factor erythroid 2-related factor 2 and Kelch-like ECH-associated protein 1 pathways. These signalling pathways collaborate with cell death mechanisms, which include apoptosis, necroptosis and autophagy, to determine cell fate. Extensive research has been carried out in our laboratory to investigate these signal transduction and cell death mechanistic pathways in CRC. This review summarizes CRC pathogenesis and the related cell death and signal transduction pathways.

9.
Biochem Biophys Res Commun ; 503(3): 1723-1731, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30075845

RESUMEN

Chemotherapeutic drugs exert systemic toxicity in lung cancer cells and therefore novel treatment strategies are warranted. Epigallocatechin 3-gallate (EGCG), though possessing beneficial effects in alleviating cancer, its effect has been limited due to ineffective systemic delivery, toxicity and bioavailability. To attain the maximum therapeutic response of EGCG, we have synthesized bovine serum albumin (BSA) encapsulated magnetite nanoparticle (MNPs) loaded with EGCG (nano EGCG). The synthesized nano EGCG was characterized using HR-TEM, XRD and FT-IR. Cytotoxicity analysis of BSA-MNP and nano EGCG using flow cytometry was evaluated in lung adenocarcinoma A549 cells. The effect of native and nano EGCG modulating apoptosis and Nrf2/Keap1 signaling was analysed. Nano EGCG exhibited increased ROS/RNS levels and decreased mitochondrial membrane potential, as evaluated by DCFH and JC1 staining, respectively. Expression of pro-apoptotic Bcl-2 family proteins (Bcl-2, Bax, Bak, Bim and Puma) was evaluated. This study demonstrates that native and nano EGCG induces apoptosis through the involvement of ROS leading to loss in mitochondrial membrane potential. EGCG exhibited an increased expression of Nrf2 and Keap1 that could regulate apoptosis in A549 cells. This study, for the first time reveals the potential of BSA-MNPs loaded EGCG as drug target and renders better efficacy against lung cancer cells.


Asunto(s)
Anticarcinógenos/farmacología , Apoptosis/efectos de los fármacos , Catequina/análogos & derivados , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células A549 , Anticarcinógenos/química , Catequina/química , Catequina/farmacología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Tamaño de la Partícula , Transducción de Señal/efectos de los fármacos , Propiedades de Superficie
10.
Basic Clin Pharmacol Toxicol ; 123(2): 122-129, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29394529

RESUMEN

The respiratory disease pulmonary fibrosis (PF), which is characterized by scar formation throughout the lung, imposes a serious health burden. No effective drug without side effects has been proven to prevent this fatal lung disease. In this context, this study was undertaken to elucidate the protective effect of celastrol, a quinine methide pentacyclic triterpenoid from a Chinese medicinal plant 'thunder god vine' against bleomycin (BLM)-induced PF. We also attempted to study how the cytokine transforming growth factor-ß (TGF-ß) stimulates fibrosis through the induction of epithelial-mesenchymal transition (EMT) and the role of celastrol in regulating EMT. TGF-ß (5 ng/ml) was administered to human alveolar epithelial adenocarcinoma A549 cells to induce fibrotic response in cells. Induction of EMT was analysed in cells through morphological analysis and expression of epithelial and mesenchymal markers by Western blotting. Bleomycin at a concentration of 3 U/Kg b.w was used to induce fibrosis in adult male rat lungs. Celastrol (5 mg/kg b.w) was given to rats twice a week after BLM administration for a period of 28 days. Western blot and immunofluorescence analyses were performed with lung tissue sample to find out the potential of celastrol in regulating EMT during the progression of fibrosis. TGF-ß induces EMT in A549 cells as demonstrated by changes in epithelial cell morphology and expression of epithelial and mesenchymal marker proteins. The expressions of epithelial marker proteins E-cadherin and claudin were found to be reduced in the BLM-induced group of rats. Expression of mesenchymal markers, such as N-cadherin, snail, slug, vimentin and ß-catenin, was enhanced in BLM-induced rat lungs. Celastrol reverts these cellular changes in rat lungs, and it was found that celastrol regulates EMT through the inhibition of heat shock protein 90 (HSP 90). Together, the results indicate that EMT is a crucial phenomenon for the progression of fibrosis, and celastrol provides protection against PF through the regulation of EMT.


Asunto(s)
Medicamentos Herbarios Chinos/uso terapéutico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Triterpenos/uso terapéutico , Células A549 , Animales , Bleomicina/toxicidad , Cadherinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Triterpenos Pentacíclicos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Ratas , Ratas Wistar , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Tripterygium/química
11.
Pulm Pharmacol Ther ; 45: 47-61, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28389259

RESUMEN

Pulmonary fibrosis (PF) is a chronic pulmonary disease of unknown cause with high mortality. Autophagy is an important homeostatic process that decides the fate of cells under stress conditions. This study is aimed to investigate whether impaired autophagic activity leads to fibrosis and pharmacological induction of autophagy provides protection against bleomycin (BLM)-induced PF. A single dose of BLM (3 U/kg body weight) was administered intratracheally to induce fibrosis in rats. Celastrol, a triterpenoid (5 mg/kg body weight, intraperitoneally) was given in every 81 h for a period of 28 days. Western blot and Confocal microscopic analysis of rat lung tissue samples revealed that celastrol induces autophagy in BLM-induced rats. Transmission electron microscopic analysis supports the above findings. Celastrol increased the expressions of Beclin 1 and Vps 34, promoted the up-regulation of Atg5-Atg12-16 formation and enhanced the lipidation of LC3I to LC3II suggesting induction of autophagy by celastrol provide protection against lung fibrosis. Further, we revealed that celastrol activates autophagy by inhibiting PI3K/Akt mediated mTOR expression. In addition, we show evidences that lack of autophagy leads to accumulation of p62, an autophagy adaptor protein that is degraded by celastrol. This study helps to describe the importance of autophagic cell death as a possible therapeutic target against lung fibrosis, and celastrol as a potential candidate for the treatment options for PF.


Asunto(s)
Autofagia/efectos de los fármacos , Fibrosis Pulmonar/prevención & control , Proteína Sequestosoma-1/metabolismo , Triterpenos/farmacología , Animales , Bleomicina/toxicidad , Western Blotting , Masculino , Microscopía Confocal , Microscopía Electrónica de Transmisión , Triterpenos Pentacíclicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fibrosis Pulmonar/fisiopatología , Ratas , Ratas Wistar , Serina-Treonina Quinasas TOR/genética
12.
Chem Biol Interact ; 246: 52-62, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26768587

RESUMEN

Pulmonary fibrosis (PF) is characterized by excessive accumulation of extracellular matrix components in the alveolar region which distorts the normal lung architecture and impairs the respiratory function. The aim of this study is to evaluate the anti-fibrotic effect of celastrol, a quinine-methide tri-terpenoid mainly found in Thunder God Vine root extracts against bleomycin (BLM)-induced PF through the enhancement of antioxidant defense system. A single intratracheal instillation of BLM (3 U/kg.bw) was administered in rats to induce PF. Celastrol (5 mg/kg) was given intraperitoneally, twice a week for a period of 28 days. BLM-induced rats exhibits declined activities of enzymatic and non-enzymatic antioxidants which were restored upon treatment with celastrol. BLM-induced rats show increased total and differential cell counts as compared to control and celastrol treated rats. Histopathological analysis shows increased inflammation and alveolar damage; while assay of hydroxyproline and Masson's trichrome staining shows an increased collagen deposition in BLM-challenged rats that were decreased upon celastrol treatment. Celastrol also reduces inflammation in BLM-induced rats as evidenced by decrease in the expressions of mast cells, Tumor necrosis factor-alpha (TNF- α) and matrix metalloproteinases (MMPs) 2 and 9. Further, Western blot analysis shows that celastrol is a potent inducer of NF-E2-related factor 2 (Nrf2) and it restores the activities of Phase II enzymes such as hemoxygenase-1 (HO-1), glutathione-S-transferase (GSTs) and NADP(H): quinine oxidoreductase (NQO1) which were declined upon BLM administration. The results of this study show evidence on the protective effect of celastrol against BLM-induced PF through its antioxidant and anti-fibrotic effects.


Asunto(s)
Antioxidantes/metabolismo , Bleomicina/farmacología , Colágeno/biosíntesis , Factor 2 Relacionado con NF-E2/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Triterpenos/farmacología , Animales , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Triterpenos Pentacíclicos , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/patología , Ratas , Ratas Wistar , Triterpenos/uso terapéutico
13.
Can J Physiol Pharmacol ; 94(1): 65-71, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26544788

RESUMEN

Naringin (4',5,7-trihydroxy-flavonone-7-rhamnoglucoside), a flavonone present in grapefruit, has recently been reported to protect against neurodegeration, induced with 3-nitropropionic acid (3-NP), through its antioxidant, anti-inflammatory, and antiapoptotic properties. This study used a rat model of 3-NP-induced neurodegeneration to investigate the neuroprotective effects of naringin exerted by modulating the expression of matrix metalloproteinases and glial fibrillary acidic protein. Neurodegeneration was induced with 3-NP (10 mg/kg body mass, by intraperitoneal injection) once a day for 2 weeks, and induced rats were treated with naringin (80 mg/kg body mass, by oral gavage, once a day for 2 weeks). Naringin ameliorated the motor abnormalities caused by 3-NP, and reduced blood-brain barrier dysfunction by decreasing the expression of matrix metalloproteinases 2 and 9, along with increasing the expression of the tissue inhibitors of metalloproteinases 1 and 2 in 3-NP-induced rats. Further, naringin reduced 3-NP-induced neuroinflammation by decreasing the expression of nuclear factor-kappa B and glial fibrillary acidic protein. Thus, naringin exerts protective effects against 3-NP-induced neurodegeneration by ameliorating the expressions of matrix metalloproteinases and glial fibrillary acidic protein.


Asunto(s)
Flavanonas/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/prevención & control , Fármacos Neuroprotectores/farmacología , Animales , Conducta Animal/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , FN-kappa B/metabolismo , Degeneración Nerviosa/inducido químicamente , Nitrocompuestos/toxicidad , Propionatos/toxicidad , Ratas , Ratas Wistar , Inhibidores Tisulares de Metaloproteinasas/metabolismo
14.
J Nutr Biochem ; 26(12): 1527-39, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26411262

RESUMEN

Dietary flavonoids have been suggested to promote brain health by protecting brain parenchymal cells. Recently, understanding the possible mechanism underlying neuroprotective efficacy of flavonoids is of great interest. Given that fisetin exerts neuroprotection, we have examined the mechanisms underlying fisetin in regulating Aß aggregation and neuronal apoptosis induced by aluminium chloride (AlCl3) administration in vivo. Male Swiss albino mice were induced orally with AlCl3 (200 mg/kg. b.wt./day/8 weeks). Fisetin (15 mg/Kg. b.wt. orally) was administered for 4 weeks before AlCl3-induction and administered simultaneously for 8 weeks during AlCl3-induction. We found aggregation of Amyloid beta (Aß 40-42), elevated expressions of Apoptosis stimulating kinase (ASK-1), p-JNK (c-Jun N-terminal Kinase), p53, cytochrome c, caspases-9 and 3, with altered Bax/Bcl-2 ratio in favour of apoptosis in cortex and hippocampus of AlCl3-administered mice. Furthermore, TUNEL and fluoro-jade C staining demonstrate neurodegeneration in cortex and hippocampus. Notably, treatment with fisetin significantly (P<0.05) reduced Aß aggregation, ASK-1, p-JNK, p53, cytochrome c, caspase-9 and 3 protein expressions and modulated Bax/Bcl-2 ratio. TUNEL-positive and fluoro-jade C stained cells were also significantly reduced upon fisetin treatment. We have identified the involvement of fisetin in regulating ASK-1 and p-JNK as possible mediator of Aß aggregation and subsequent neuronal apoptosis during AlCl3-induced neurodegeneration. These findings define the possibility that fisetin may slow or prevent neurodegneration and can be utilised as neuroprotective agent against Alzheimer's and Parkinson's disease.


Asunto(s)
Compuestos de Aluminio/efectos adversos , Apoptosis , Corteza Cerebral/patología , Cloruros/efectos adversos , Flavonoides/química , Hipocampo/patología , Neuronas/patología , Administración Oral , Cloruro de Aluminio , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Corteza Cerebral/efectos de los fármacos , Citocromos c/metabolismo , Dieta , Flavonoles , Hipocampo/efectos de los fármacos , MAP Quinasa Quinasa Quinasa 5/metabolismo , Masculino , Ratones , Enfermedades Neurodegenerativas/patología , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
15.
Brain Res ; 1620: 42-56, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25935694

RESUMEN

Neuronal apoptosis occurs as a sequel of oxidative stress associated with various neuropathies. In this study, we have investigated the protective effect of farnesol, a sequisterpene on lipopolysaccharide (LPS) induced neurodegeneration through modulation of intrinsic apoptotic cascade in the cortex and hippocampus of Swiss albino mice. Intraperitoneal (i.p.) injection of LPS (250 µg/kg b.wt. for 7 days) resulted in elevated levels of lipid peroxidation, protein carbonyls and 8-Hydroxydeoxyguanosine (8OHdG), with subsequent depletion in the antioxidant status and severe histological aberrations. These anomalies were accompanied by increased expressions of pro-apoptotic Bax, caspase-3 and p53 with decrease in anti-apoptotic Bcl-2. Farnesol treatment (100mg/kg b.wt.) ameliorated LPS-induced oxidative stress by enhancing the antioxidant defense system as evident from the increased levels of SOD, CAT, GSH and GST and exhibited protected cellular morphology manifested from histopathological and nissl staining analyses. Farnesol treatment also reduced the expulsion of cytochrome c from mitochondria and downregulated caspase 3 activation as revealed by immunoblot analysis. Furthermore, farnesol treatment reduced the expression of Bax and antagonized LPS-induced decrease in anti-apoptotic Bcl-2. Results of this study show that farnesol exerts neuroprotective effect by regulating intrinsic apoptotic cascade through its antioxidant effect during LPS-induced neurodegeneration.


Asunto(s)
Apoptosis/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Farnesol/farmacología , Degeneración Nerviosa/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Animales , Antioxidantes/metabolismo , Apoptosis/fisiología , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Escherichia coli , Lipopolisacáridos , Masculino , Ratones , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Distribución Aleatoria
16.
Clin Exp Pharmacol Physiol ; 42(8): 849-59, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26010495

RESUMEN

Pulmonary fibrosis (PF) poses a huge burden to the patients and society due to lack of an effective treatment drug. Activation of fibrocyte, fibroblast and myofibroblasts are important steps in the development of PF. Targeting this common pathway with natural chemicals may lead to the development of new drug regimens for PF treatment. In this study, PF was induced in male Wistar rats by intratracheal administration of Bleomycin (BLM). Epigallocatechin gallate (EGCG) was administered to one of the groups of rats to test its efficacy against the development of PF. Bleomycin-induction resulted in significant elevation of matrix metalloproteinase (MMP)-2 and -9 expression, increased RNA and protein expression of transforming growth factor (TGF)-ß1, Smads and alpha-smooth muscle actin (α-SMA). EGCG treatment normalized the BLM induced aberrations in these rats. The protective role of EGCG was also validated in vitro using the WI-38 fibroblast cell line. TGF-ß1 incubated cells exhibited increased fibroblast proliferation and hydroxyproline levels with a concomitant decrease in the expression of MMPs 2 and 9. An increase in protein expression levels of p-Smad, α-SMA and type I collagen (COL1A) was also exhibited by fibroblasts upon TGF-ß1 incubation. Simultaneous treatment of EGCG to WI-38 cells significantly decreased these protein expressions alongside normalizing the MMPs expression. The study revealed that EGCG inhibited fibroblast activation and collagen accumulation by inhibiting TGF-ß1 signalling and thus can be considered as an effective drug against PF.


Asunto(s)
Catequina/análogos & derivados , Colágeno/biosíntesis , Fibroblastos/citología , Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Bleomicina/farmacología , Catequina/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Ratas , Ratas Wistar
17.
J Mol Med (Berl) ; 93(9): 1015-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25877860

RESUMEN

UNLABELLED: Idiopathic pulmonary fibrosis (IPF) is a fibroproliferative lung disorder of unknown aetiology. Transforming growth factor-ß1 (TGF-ß1)-mediated Smad and non-Smad signaling cascades are considered as central players in accelerating pulmonary fibrosis. We earlier reported berberine's amelioration against TGF-ß1-mediated pro-fibrotic effects in bleomycin-induced pulmonary fibrosis. The present study aimed to determine the regulatory role of berberine on abrogated Smad 2/3 and FAK-dependent PI3K/Akt-mTOR signaling cascades in bleomycin-induced pulmonary fibrosis. Male Wistar rats were subjected to single intratracheal instillation of bleomycin (2.5 U/kg) on day 0, and berberine treatments were provided in either preventive or therapeutic modes, respectively. Berberine mitigated the elevated expression of fibrotic markers, α-smooth muscle actin (α-SMA), fibronectin, collagens I and III and reversed bleomycin-induced ultrastructural alterations in the lungs. Berberine inhibited the bleomycin-induced raise in p-Smad 2/3 and enhanced Smad 7 expression. Berberine blocked the activation of FAK and PI3K/Akt against bleomycin-induced dysregulation, with subsequent raise in PTEN expression. In addition, by inhibiting p-mTOR, berberine stimulated autophagy as evidenced by increase in Beclin-1, LC3-II levels with enhanced autophagosome formation. Cumulatively, through targeted inhibition of dysregulated Smad and FAK-dependent PI3K/Akt-mTOR signaling axis, berberine attenuated the fibrotic insults of bleomycin. KEY MESSAGE: Berberine inhibits Smad 2/3 activation and enhances Smad 7 in bleomycin-induced rat lungs. Bleomycin-induced activation of FAK is inhibited by berberine. Berberine inhibits bleomycin-induced activation of PI3K/Akt cascade. Berberine inhibits mTOR activation to enhance autophagy and suppresses fibrotic markers.


Asunto(s)
Autofagia/efectos de los fármacos , Berberina/uso terapéutico , Fibrosis Pulmonar/tratamiento farmacológico , Proteínas Smad/metabolismo , Animales , Bleomicina/toxicidad , Immunoblotting , Inmunohistoquímica , Masculino , Microscopía Confocal , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fibrosis Pulmonar/inducido químicamente , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
18.
Biochem Biophys Res Commun ; 461(2): 314-20, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25881504

RESUMEN

Chronic inflammation is one of the remarkable etiologic factors for various human ailments including cancer. The well known hypothesis is that persistent inflammation in colon can increase the risk of colorectal cancer (CRC). In this study, a pharmacological evaluation of carvacrol, a phenolic monoterpene constituent of essential oils produced from aromatic plant Oreganum vulgarea sp. on colitis associated colon cancer (CACC) induced by 1,2 Dimethylhydrazine (DMH) and dextran sodium sulfate (DSS) in male Fischer 344 rat model was studied. F344 rats were given three subcutaneous injections of DMH (40 mg/kg body wt) in the first week and were given free access to drinking water containing 1% DSS for the next one week followed by 7-14 days of water as three cycles. Carvacrol was administrated before and after tumor induction at a concentration of 50 mg/kg body weight (o.p). Carvacrol treated groups promotes the endogenous antioxidant system and suppress the inflammation in DMH/DSS induced animals. An increased antioxidant status and restoration of histological lesions in the inflamed colonic mucosa was observed in carvacrol treated rats. This effect was confirmed biochemically by reducing free-radical accumulation and suppressing expression of pro-inflammatory mediators. In this study, Carvacrol significantly increased the anti-oxidant enzymes such as superoxide dismutase (SOD), catalase (CAT) glutathione (GSH) levels and reduced lipid peroxides (LPO), myeloperoxidase (MPO) and nitric oxide (NO) as compared to DMH/DSS induced rats. These dramatic changes facilitate the suppression of pro-inflammatory mediators such as inducible nitric oxide synthase (iNOS), and interleukin-1 beta (IL-1ß) in CACC induced rats. Taken together, these findings suggest that Carvacrol may play a beneficial role in DMH/DSS induced experimental rat model and serve as an excellent dietary antioxidant as well as anti-inflammatory agent. It may represent novel therapeutic interventions against colon cancer triggered by chronic inflammation.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Antioxidantes/uso terapéutico , Colitis/tratamiento farmacológico , Colon/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Monoterpenos/uso terapéutico , Animales , Antiinflamatorios/farmacología , Antineoplásicos Fitogénicos/farmacología , Antioxidantes/farmacología , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/patología , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/etiología , Neoplasias del Colon/patología , Cimenos , Sulfato de Dextran , Interleucina-1beta/análisis , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/patología , Monoterpenos/farmacología , Óxido Nítrico Sintasa de Tipo II/análisis , Ratas , Ratas Endogámicas F344
19.
ACS Appl Mater Interfaces ; 7(3): 1422-30, 2015 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-25552345

RESUMEN

We report a novel green chemical approach for the synthesis of blue light-emitting and water-soluble Ag subnanoclusters, using sodium cholate (NaC) as a template at a concentration higher than the critical micelle concentration (CMC) at room temperature. However, under photochemical irradiation, small anisotropic and spherically shaped Ag nanoparticles (3-11 nm) were obtained upon changing the concentration of NaC from below to above the CMC. The matrix-assisted laser desorption ionization time-of-flight and electrospray ionization mass spectra showed that the cluster sample was composed of Ag4 and Ag6. The optical properties of the clusters were studied by UV-visible and luminescence spectroscopy. The lifetime of the synthesized fluorescent Ag nanoclusters (AgNCs) was measured using a time-correlated single-photon counting technique. High-resolution transmission electron microscopy was used to assess the size of clusters and nanoparticles. A protocol for transferring nanoclusters to organic solvents is also described. Toxicity and bioimaging studies of NaC templated AgNCs were conducted using developmental stage zebrafish embryos. From the survival and hatching experiment, no significant toxic effect was observed at AgNC concentrations of up to 200 µL/mL, and the NC-stained embryos exhibited blue fluorescence with high intensity for a long period of time, which shows that AgNCs are more stable in living system.


Asunto(s)
Embrión no Mamífero/química , Imagen Molecular/instrumentación , Plata/química , Colato de Sodio/química , Pez Cebra/embriología , Animales , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/efectos de la radiación , Fluorescencia , Luz , Nanopartículas del Metal/química , Nanopartículas del Metal/toxicidad , Plata/toxicidad , Colato de Sodio/toxicidad
20.
Mol Cell Biochem ; 399(1-2): 303-13, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25355159

RESUMEN

Chemoprevention is regarded as one of the most promising and realistic approaches in the prevention of human cancer. Ellagic acid (EA) has been known for its chemopreventive activity against various cancers and numerous investigations have shown its apoptotic activity both in vivo and in vitro. The present study was focused to elucidate the anticancerous effect and the mode of action of EA against HCT-15 colon adenocarcinoma cells. Cell viability was assessed using trypan blue assay at different concentrations. EA also promoted cell cycle arrest substantially at G2/M phase in HCT-15 cells. The activities of alkaline phosphatase and lactate dehydrogenase were decreased upon EA treatment, which shows the antiproliferative and the cytotoxic effects, respectively. The production of reactive oxygen intermediates, which were examined by 2,7-dichlorodihydrofluorescein diacetate (H2DCF-DA), increased with time, after treatment with EA. In further studies, EA inhibited proliferation-associated markers proliferating cell nuclear antigen and cyclin D1. The induction of apoptosis was accompanied by a strong inactivation of phosphatidylinositol 3-kinase (PI3K)/Akt pathway by EA. The expression of PI3K and pAkt was down-regulated in EA-treated cells, compared to normal cells. Further, EA promoted the expression of Bax, caspase-3, and cytochrome c, and suppression of Bcl-2 activity in HCT-15 cells that was determined by western blot analysis. Increased annexin V apoptotic cells and DNA fragmentation also accompanied EA-induced apoptosis. In conclusion, EA increased the production of ROS, decreased cell proliferation, and induced apoptosis in HCT-15 cells, and thus can be used as an agent against colon cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Proliferación Celular/efectos de los fármacos , Ácido Elágico/farmacología , Transducción de Señal , Adenocarcinoma/tratamiento farmacológico , Fosfatasa Alcalina/metabolismo , Línea Celular Tumoral , Forma de la Célula , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Ciclina D1/metabolismo , Fragmentación del ADN , Ensayos de Selección de Medicamentos Antitumorales , Puntos de Control de la Fase G2 del Ciclo Celular , Humanos , L-Lactato Deshidrogenasa/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA