Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Biomed Pharmacother ; 109: 2182-2191, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30551475

RESUMEN

This work focused on characterizing hydrophilic fractions of Clematis flammula (CFl). The data here clearly demonstrated that hydrolate fractions act as a free radical scavengers and inhibited proliferation of different cell lines in a time- and concentration-dependent manner, transwell, and with a significant cytotoxic effect. Treating cells with CFl had the effect of suppressing cell growth attenuated by ROS generation in colonic carcinoma. Moreover, CFl in HCT116 cells suppressed survival, proliferation, invasion, angiogenesis and metastasis in vitro by inhibiting gene expression. Following CFl treatment, caspases and PARP cleavage were detected. The up- and down-regulated genes obtained from the WBA of the effect of CFl showed that several biological processes were associated with apoptosis and induction of G1 cell cycle arrest. CFl synergizes the effect of TRAIL by down-regulating the expression of cell survival proteins involved in apoptosis compared to cells treated with CFl or TRAIL alone. Our findings showed that CFl sensitizes apoptosis in TRAIL-resistant cells by activating MAPKs, SP1, and CHOP, that induced DR5 expression. Overall, our data showed that CFl is a promising antitumor agent through kinases and transcription factor induction, both of which are required to activate TRAIL receptors. Colon inflammation induced by LPS was inhibited by CFl hydrolate.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Clematis , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Extractos Vegetales/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Antineoplásicos Fitogénicos/aislamiento & purificación , Antineoplásicos Fitogénicos/farmacología , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HL-60 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Células MCF-7 , Ratones , Componentes Aéreos de las Plantas , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología
3.
Oncotarget ; 9(64): 32305-32320, 2018 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-30190788

RESUMEN

This study focused on characterizing the Hydrophobic and Hydrophilic fractions of Teucrium alopecurus in the context of cancer prevention and therapy. The goal was also to elucidate the molecular mechanisms involved and to determine its efficacy against cancer by triggering apoptosis and suppressing tumorigenesis in human colon cancer. The data here clearly demonstrated that oily fractions of Teucrium alopecurus act as free radical scavengers, antibacterial agent and inhibited the proliferation of HCT-116, U266, SCC4, Panc28, KBM5, and MCF-7 cells in a time- and concentration-dependent manner. The results of live/dead and colony formation assays further revealed that Teucrium essential oil has the efficacy to suppress the growth of colon carcinoma cells. In addition, essential oil of Teucrium alopecurus induced apoptosis, as indicated by cleavage of caspases-3, -8, and -9 and poly-adenosine diphosphate ribose polymerase. Moreover, Teucrium alopecurus essential oil suppressed gene expression involved in survival, proliferation, invasion, angiogenesis, and metastasis in human colon cancer cells. No sign of toxicity was detected in vivo after treatment with increasing concentrations of essential oil. Oral administration of T.alopecurus inhibited LPS-induced colon inflammation. This anticancer property of this specie Teucrium alopecurus fractions could be due to their phenolic and/or sesquiterpene content (d-limonene, α-Bisabolol, Humulene, Thymol, and (+)-epi-Bicyclosesquiphellandrene). Hence our study reveals the anticancer activity of Teucrium alopecurus oil mediated through the suppression of cell growth, cell proliferation, and the induction of apoptosis of cancer cells. Thus, it has potential to be developed as an anticancer agent; however more in vitro and in vivo studies are warranted.

4.
Cell Death Discov ; 4: 15, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29531812

RESUMEN

Stress responses are critical for estrogen (E2)-induced apoptosis in E2-deprived breast cancer cells. Nuclear factor-kappa B (NF-κB) is an important therapeutic target to prevent stress responses in chronic inflammatory diseases including cancer. However, whether E2 activates NF-κB to participate in stress-associated apoptosis in E2-deprived breast cancer cells is unknown. Here, we demonstrated that E2 differentially modulates NF-κB activity according to treatment time. E2 initially has significant potential to suppress NF-κB activation; it completely blocks tumor necrosis factor alpha (TNFα)-induced activation of NF-κB. We found that E2 preferentially and constantly enhances the expression of the adipogenic transcription factor CCAAT/enhancer binding protein beta (C/EBPß), which is responsible for the suppression of NF-κB activation by E2 in MCF-7:5C cells. Interestingly, NF-κB p65 DNA-binding activity is increased when E2 is administered for 48 h, leading to the induction of TNFα and associated apoptosis. Blocking the nuclear translocation of NF-κB can completely prevent the induction of TNFα and apoptosis induced by E2. Further examination revealed that protein kinase RNA-like endoplasmic reticulum kinase (PERK), a stress sensor of unfolded protein response (UPR), plays an essential role in the late activation of NF-κB by E2. This modulation between PERK and NF-κB is mainly mediated by a stress responsive transcription factor, transducer and activator of transcription 3 (STAT3), independently of the classic canonical IκBα signaling pathway. Thus, inhibition of PERK kinase activity completely blocks the DNA binding of both STAT3 and NF-κB, thereby preventing induction of NF-κB-dependent genes and E2-induced apoptosis. All of these findings suggest that PERK is a key regulator to convey stress signals from the endoplasmic reticulum to the nucleus and illustrate a crucial role for the novel PERK/STAT3/NF-κB/TNFα axis in E2-induced apoptosis in E2-deprived breast cancer cells.

5.
Biomed Pharmacother ; 95: 1876-1885, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28968948

RESUMEN

Teucrium alopecurus is an endemic plant limited to southern Tunisia. In the present study, the chemical composition, anticancer and nuclear factor-κB (NF-κB) inhibitory effects of Teucrium alopecurus leaf essential oil was investigated. The analysis of Teucrium alopecurus (TA-1) with Gas Chromatography-Mass Spectrometry (GC/MS) showed that α-Bisabolol, (+)-epi-Bicyclosesquiphellandrene and α-Cadinol, were found in relatively high amounts (16.16%, 15.40% and 8.52%, respectively). Cell viability was determined by 3-(4-5-dimethylthiazol-2-yl) 2-5-diphenyl-tetrazolium (MTT) assay. Cell cycle and apoptosis assay were determined by flow cytometry. TA-1 functions as an anticancer agent by triggering apoptosis potentiated by chemotherapeutic agents and TNF in human myeloid leukemia cells (KBM5) through a mechanism involving poly(ADP-ribose) polymerase (PARP) cleavage and initiator and effector caspases activation. Moreover, electrophoretic mobility shift assay (EMSA) revealed that TA-1 downregulated nuclear localization of NF-κB and its phosphorylation induced by TNF-α and this, allows the suppression of the degradation and phosphorylation of IκB and the inhibition of the phosphorylation of p65 phosphorylation and the p50-p65 heterodimer nuclear translocation, causing attenuation of NF-κB-regulated antiapoptotic (Survivin, Bcl-2, c-IAP1/2, Bcl-xL, Mcl-1, and cFLIP), invasion (ICAM1), metasatsis (MMP-9), and angiogenesis (VEGF) gene expression in KBM5; and finally reporter gene expression. Furthermore, treatment with essential oil and TNF-α suppressed the NF-κB DNA binding activity. Finally, the activation of nuclear factor-κB induced by different plasmids (TNFR1, TRADD, TRAF2, NIK, TAK1/TAB1, and IKKß) was inhibited following treatment with TA-1. Overall, TA-1 inhibits NF-κB activation and further growth and proliferation of cancer cells.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Aceites Volátiles/farmacología , Terpenos/farmacología , Teucrium/química , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/farmacología , Antineoplásicos Fitogénicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Citometría de Flujo , Cromatografía de Gases y Espectrometría de Masas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/metabolismo , Leucemia Mieloide/tratamiento farmacológico , FN-kappa B/metabolismo , Aceites Volátiles/aislamiento & purificación , Hojas de la Planta , Terpenos/aislamiento & purificación , Factor de Necrosis Tumoral alfa/metabolismo , Túnez
6.
Phytomedicine ; 34: 14-20, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28899496

RESUMEN

BACKGROUND: For centuries, agents derived from natural sources (mother nature), especially plants have been the primary source of medicine. Neem, also referred to as Azadirachta indica is one such plant that has been so named because it provides freedom from all diseases, and used for thousands of years in Indian and African continents. Different parts of the plant including flowers, leaves, seeds and bark have been used to treat both acute and chronic human diseases; and used as insecticide; antimicrobial, larvicidal, antimalarial, antibacterial, antiviral, and spermicidal. PURPOSE: What is there in neem and how it manifests its wide variety of effects is the focus of this review. How neem and its constituents modulate various cellular pathways is discussed. The animal and human studies carried out with neem and its constituents is also discussed. CONCLUSION: Over 1000 research articles published on neem has uncovered over 300 structurally diverse constituents, one third of which are limonoids including nimbolide, azadarachtin, and gedunin. These agents manifest their effects by modulating multiple cell signaling pathways.


Asunto(s)
Azadirachta/química , Limoninas/química , Fitoquímicos/farmacología , Extractos Vegetales/farmacología , Animales , Ensayos Clínicos como Asunto , Flores/química , Humanos , Estructura Molecular , Fitoquímicos/química , Corteza de la Planta/química , Hojas de la Planta/química , Semillas/química
7.
Phytomedicine ; 34: 171-181, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28899500

RESUMEN

BACKGROUND: While the anti-inflammatory and anticancer potential of curcumin, which is derived from turmeric (Curcuma longa), has been studied extensively, very little is known about Calebin A, another novel compound from the same source. PURPOSE: To determine whether Calebin A exhibits anti-inflammatory and anticancer potential. METHODS: We examined the anti-inflammatory potential of Calebin A by DNA binding of NF-κB. Anticancer properties of Calebin were determined by MTT and FACS analysis and NF-κB regulated expression of proteins was assessed by western blotting. RESULTS: Calebin A suppressed NF-κB activation induced by various stimuli. This inhibition of NF-κB activation was mediated through the suppression of direct binding of NF-κB/p65 to the DNA. This inhibitory effect was reversed by a reducing agent, and mutation of the Cys38 of p65 to serine abolished the effect of Calebin A on this binding. Suppression of NF-κB activation by Calebin A resulted in the down-regulation of the expression of proteins involved in tumor cell survival, proliferation, inflammation, and metastasis. Furthermore, Calebin A inhibited proliferation and induced apoptosis in a wide variety of tumor cells, as examined by various assays. It enhanced apoptosis induced by chemotherapeutic agents. CONCLUSION: Our results demonstrate that Calebin A inhibits NF-κB activation pathway through interaction with p65 and potentiates apoptosis in cancer cells; thus, it has potential in the treatment of cancer. However, further in vivo studies are warranted to define its anti-inflammatory and anticancer potential.


Asunto(s)
Antiinflamatorios/farmacología , Antineoplásicos/farmacología , Cinamatos/farmacología , Curcuma/química , Monoterpenos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Humanos , Fosforilación , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo
8.
Front Pharmacol ; 8: 871, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29311914

RESUMEN

Extensive research within last two decades has indicated that curcumin extracted from turmeric (Curcuma longa), exhibits anticancer potential, in part through the modulation of inflammatory pathways. However, the residual antitumor activity of curcumin-free turmeric (CFT) relative to curcumin or turmeric is not well-understood. In the present study, therefore, we determined activities of these agents in both in vitro and in vivo models of human HCT-116 colorectal cancer (CRC). When examined in an in vitro antiproliferative, clonogenic or anti-inflammatory assay system, we found that curcumin was highly active whereas turmeric and CFT had relatively poor activity against CRC cells. However, when examined in vivo at an oral dose of either 100 or 500 mg/kg given to nude mice bearing CRC xenografts, all three preparations of curcumin, turmeric, and CFT similarly suppressed the growth of the xenograft. The effect of CFT on suppression of tumor growth was dose-dependent, with 500 mg/kg tending to be more effective than 100 mg/kg. Interestingly, 100 mg/kg curcumin or turmeric was found to be more effective than 500 mg/kg. When examined in vivo for the expression of biomarkers associated with cell survival (cIAP-1, Bcl-2, and survivin), proliferation (Ki-67 and cyclin D1) and metastasis (ICAM-1 and VEGF), all were down-modulated. These agents also suppressed inflammatory transcription factors (NF-κB and STAT3) in tumor cells. Overall, our results with CFT provide evidence that turmeric must contain additional bioactive compounds other than curcumin that, in contrast to curcumin, exhibit greater anticancer potential in vivo than in vitro against human CRC. Moreover, our study highlights the fact that the beneficial effects of turmeric and curcumin in humans may be more effectively realized at lower doses, whereas CFT could be given at higher doses without loss in favorable activity.

9.
Cancer Lett ; 387: 95-105, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-27037062

RESUMEN

Extensive research over the past half a century indicates that reactive oxygen species (ROS) play an important role in cancer. Although low levels of ROS can be beneficial, excessive accumulation can promote cancer. One characteristic of cancer cells that distinguishes them from normal cells is their ability to produce increased numbers of ROS and their increased dependence on an antioxidant defense system. ROS are produced as a byproduct intracellularly by mitochondria and other cellular elements and exogenously by pollutants, tobacco, smoke, drugs, xenobiotics, and radiation. ROS modulate various cell signaling pathways, which are primarily mediated through the transcription factors NF-κB and STAT3, hypoxia-inducible factor-1α, kinases, growth factors, cytokines and other proteins, and enzymes; these pathways have been linked to cellular transformation, inflammation, tumor survival, proliferation, invasion, angiogenesis, and metastasis of cancer. ROS are also associated with epigenetic changes in genes, which is helpful in diagnosing diseases. This review considers the role of ROS in the various stages of cancer development. Finally, we provide evidence that nutraceuticals derived from Mother Nature are highly effective in eliminating cancer cells.


Asunto(s)
Antioxidantes/uso terapéutico , Suplementos Dietéticos , Neoplasias/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos , Neoplasias/metabolismo
10.
Front Microbiol ; 7: 1748, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27877160

RESUMEN

The aqueous leaf extract of Jatropha curcas was used for the synthesis of silver nanoparticles (Jc-AgNps) which were further evaluated for its antibacterial potential against food borne pathogens. J. curcas leaf extract could synthesize stable silver nanoparticles (Zeta potential: -23.4 mV) with absorption band at 430 nm. Fourier transform infrared spectroscopy indicated various biological compounds responsible for capping and stabilizing Jc-AgNps in suspension, while the presence of silver was authenticated by scanning electron microscopy (SEM) equipped with energy-dispersive X-ray. Jc-AgNps were confirmed to be uniform in shape, size and behavior through dynamic light scattering, transmission electron microscopy (TEM), X-ray diffraction, SEM, and atomic force microscopy (AFM) analysis. To investigate the antibacterial activity, disk diffusion and microplate dilution assays were performed and zone of inhibition (ZOI) as well as minimum inhibitory/bactericidal concentrations (MIC/MBCs) were evaluated against selected bacterial strains. Overall results showed that Escherichia coli (ZOI: 23 mm, MBC: 0.010 mg/ml) was the most sensitive organism, whereas Staphylococcus aureus (ZOI: 14.66 mm, MBC: 0.041 mg/ml) and Salmonella enterica (ZOI: 16.66 mm, MBC: 0.041 mg/ml) were the least sensitive against Jc-AgNps. The detailed microscopic investigations using SEM, TEM, and AFM were performed to understand the antibacterial impacts of Jc-AgNps against Listeria monocytogenes. SEM and TEM analysis showed the clear deformation and disintegration of treated L. monocytogenes cells, whereas AFM established a decrease in the height and cell surface roughness (root mean square value) in the treated L. monocytogenes.

11.
Biomed Environ Sci ; 29(9): 639-649, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27806746

RESUMEN

OBJECTIVE: This study was conducted to determine the histopathological and biochemical effects of Thymus algeriensis essential oil (TEO) on hydrogen peroxide (H2O2)-induced oxidative stress in liver and kidney tissues of rats. METHODS: Rats were treated in six groups and were exposed for 2 weeks to low (LD; 100 µmol/L) and high doses (HD; 1 mmol/L) of H2O2 in the presence or absence of TEO (180 mg/kg). Liver and kidney atrophy was measured by using biochemical and histopathological assays. RESULTS: Our study demonstrated that H2O2 induced liver and kidney atrophy, as evidenced by the significant elevation of serum aminotransferase, urea, and creatinine levels compared with those in the control rats. Urea levels were estimated by evaluating the activity of serum urease that hydrolyzes urea into CO2 and ammonia. However, TEO treatment significantly alleviated oxidative stress in the H2O2-induced liver and kidney toxicity model by reducing the levels of malondialdehyde concomitantly with marked elevations in superoxide dismutase, catalase, glutathione peroxidase, and glutathione S-transferase, as well as decrease in glutathione activity. CONCLUSION: Our data demonstrated that TEO protected against H2O2 toxicity by decreasing oxidant levels and DNA damage, as well as increasing antioxidant levels, indicating that TEO has a spectrum of antioxidant and DNA-protective properties.


Asunto(s)
Antioxidantes/farmacología , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Malondialdehído/metabolismo , Aceites Volátiles/farmacología , Extractos Vegetales/farmacología , Thymus (Planta)/química , Animales , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/toxicidad , Riñón/fisiología , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/fisiología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos
12.
Front Microbiol ; 7: 1394, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27708620

RESUMEN

Spices have been used since ancient times. Although they have been employed mainly as flavoring and coloring agents, their role in food safety and preservation have also been studied in vitro and in vivo. Spices have exhibited numerous health benefits in preventing and treating a wide variety of diseases such as cancer, aging, metabolic, neurological, cardiovascular, and inflammatory diseases. The present review aims to provide a comprehensive summary of the most relevant and recent findings on spices and their active compounds in terms of targets and mode of action; in particular, their potential use in food preservation and enhancement of shelf life as a natural bioingredient.

13.
Oncotarget ; 7(44): 71235-71254, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27659519

RESUMEN

Aromatase inhibitor (AI) resistance during breast cancer treatment is mimicked by MCF-7:5C (5C) and MCF-7:2A (2A) cell lines that grow spontaneously. Survival signaling is reconfigured but cells are vulnerable to estradiol (E2)-inducible apoptosis. These model systems have alterations of stress related pathways including the accumulation of endoplasmic reticulum, oxidative, and inflammatory stress that occur prior to E2-induced apoptosis. We investigated miRNA expression profiles of 5C and 2A to characterize their AI resistance phenotypes. Affymetrix GeneChip miRNA2.0 arrays identified 184 miRNAs differentially expressed in 2A and 5C compared to E2-free wild-type MCF-7:WS8. In 5C, 34 miRNAs of the DLK1-DIO3 locus and miR-31 were overexpressed, whereas miR-222 was low. TCGA data revealed poor and favorable overall survival for low miR-31 and miR-222 levels, respectively (HR=3.0, 95% CI:1.9-4.8; HR=0.3, 95% CI:0.1-0.6). Targets of deregulated miRNAs were identified using CLIP-confirmed TargetScan predictions. KEGG enrichment analyses for 5C- and 2A-specific target gene sets revealed pathways associated with cell proliferation including insulin, mTOR, and ErbB signaling as well as immune response and metabolism. Key genes overrepresented in 5C- and 2A-specific pathway interaction networks including EGFR, IGF1R and PIK3R1 had lower protein levels in 5C compared to 2A and were found to be differentially modulated by respective miRNA sets. Distinct up-regulated miRNAs from the DLK1-DIO3 locus may cause these attenuative effects as they are predicted to interact with corresponding 3' untranslated regions. These new miRNA profiles become an important regulatory database to explore E2-induced apoptotic mechanisms of clinical relevance for the treatment of resistant breast cancer.


Asunto(s)
Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , MicroARNs/fisiología , Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos , Receptores ErbB/genética , Estradiol/farmacología , Femenino , Humanos , Células MCF-7 , Transcriptoma
14.
Br J Cancer ; 115(7): 814-24, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27575851

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is one of the most common malignancies worldwide and even develops resistance to chemotherapeutic agents over time. As a result survival for patients with CRC remains poor. METHOD: We investigated both in vitro and in vivo effects of γ-tocotrienol (γ-T3) alone and in combination with capecitabine. Apoptosis and cytotoxicity assays were performed by MTT and FACS analysis, whereas expression of proteins was investigated using western blotting and immunohistochemistry. RESULTS: The γ-T3 inhibited the proliferation of CRC cells with wild-type or mutated KRAS. It also induced apoptosis, inhibited colony formation, and suppressed key regulators of cell survival, cell proliferation, invasion, angiogenesis, and metastasis. Furthermore, γ-T3 enhanced the anticancer effects of capecitabine in CRC cells. In a nude mouse xenograft model of human CRC, oral administration of γ-T3 inhibited tumour growth and enhanced the antitumour efficacy of capecitabine. Western blot and immunohistochemical analysis results indicated that expression of Ki-67, cyclin D1, MMP-9, CXCR4, NF-κB/p65, and VEGF was lower in tumour tissue from the combination treatment group. Combination treatment also downregulated NF-κB and NF-κB-regulated gene products. CONCLUSIONS: Our findings suggest that γ-T3 inhibited the growth of human CRC and sensitised CRC to capecitabine by regulating proteins linked to tumourigenesis.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antimetabolitos Antineoplásicos/uso terapéutico , Capecitabina/uso terapéutico , Cromanos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Vitamina E/análogos & derivados , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromanos/farmacología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Genes ras , Humanos , Masculino , Ratones , Ratones Desnudos , Mutación , FN-kappa B/metabolismo , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Ensayo de Tumor de Célula Madre , Vitamina E/farmacología , Vitamina E/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Curr Pharm Des ; 22(27): 4151-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27262330

RESUMEN

BACKGROUND: Spice and spice-derived compounds have been identified and explored for their health benefits since centuries. One of the spice long pepper has been traditionally used to treat chronic bronchitis, asthma, constipation, gonorrhea, paralysis of the tongue, diarrhea, cholera, malaria, viral hepatitis, respiratory infections, stomach ache, diseases of the spleen, cough, and tumors. METHODS: In this review, the evidences for the chemopreventive and chemotherapeutic potential of piperlongumine have been described. RESULTS: The active component piperlonguime has shown effective against various ailments including cancer, neurogenerative disease, arthritis, melanogenesis, lupus nephritis, and hyperlipidemic. These beneficial effects of piperlongumine is attributed to its ability to modulate several signaling molecules like reactive oxygen species, kinases, proteasome, proto-oncogenes, transcription factors, cell cycle, inflammatory molecules and cell growth and survival molecules. Piperlongumine also chemosensitizes to drugs resistant cancer cells. CONCLUSION: Overall the consumption of long peppers is therefore recommended for the prevention and treatment of various diseases including cancer, and thus piperlongumine may be a promising future candidate drug against cancer.


Asunto(s)
Dioxolanos/uso terapéutico , Neoplasias/tratamiento farmacológico , Especias , Artritis/tratamiento farmacológico , Encefalopatías/tratamiento farmacológico , Dioxolanos/química , Humanos , Hiperlipidemias/tratamiento farmacológico , Nefritis Lúpica/tratamiento farmacológico
17.
Biomed Environ Sci ; 29(4): 275-85, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27241738

RESUMEN

OBJECTIVE: We evaluate the effects of Thymus algeriensis (TEO) against hydrogen peroxide (H2O2) toxicity on body and testis weight, testis sperm count, testis lipid peroxidation, and antioxidant enzyme activities in rats. METHODS: Rats were treated with low (LD) and high dose (HD) of H2O2 (0.1 and 1 mmol/L) in the presence or absence of TEO (150 mg/kg). RESULTS: The results exhibited a significant decrease in body weight and testis weight, in total sperm number decrease (P<0.05), sperm motility and percentage of sperm viability, leading to complete arrest, in sperm flagellar beat frequency by the gavage of 1 mmol/L H2O2 compared to controls. The administration of H2O2 resulted in a significant reduction in testis GSH, GPx, CAT, SOD, and GST activity and significant increase (P<0.05) in MDA concentration compared with the untreated control animals. TEO pre-treatment protected testis from the H2O2 generated oxidative stress. These results were confirmed by histological architecture examinations. CONCLUSION: H2O2 has the ability to alter the sperm function, characteristics and development of testis. However, TEO is an efficient natural agent, which can prevent the testis from H2O2-induced oxidative damage in rats.


Asunto(s)
Peróxido de Hidrógeno/toxicidad , Estrés Oxidativo , Extractos Vegetales/farmacología , Testículo/efectos de los fármacos , Thymus (Planta)/química , Animales , Masculino , Ratas , Ratas Wistar
18.
Exp Biol Med (Maywood) ; 241(8): 783-99, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-27013544

RESUMEN

Inflammation is a part of the complex biological response of inflammatory cells to harmful stimuli, such as pathogens, irritants, or damaged cells. This inflammation has been linked to several chronic diseases including cancer, atherosclerosis, rheumatoid arthritis, and multiple sclerosis. Major biomarkers of inflammation include tumor necrosis factor, interleukins (IL)-1, IL-6, IL-8, chemokines, cyclooxygenase, 5-lipooxygenase, and C-reactive protein, all of which are regulated by the transcription factor nuclear factor-kappaB. Although examining inflammatory biomarkers in blood is a standard practice, its identification in saliva and/or urine is more convenient and non-invasive. In this review, we aim to (1) discuss the detection of these inflammatory biomarkers in urine and saliva; (2) advantages of using salivary and urinary inflammatory biomarkers over blood, while also weighing on the challenges and/or limitations of their use; (3) examine their role(s) in connection with diagnosis, prevention, treatment, and drug development for several chronic diseases with inflammatory consequences, including cancer; and (4) explore the use of innovative salivary and urine based biosensor strategies that may permit the testing of biomarkers quickly, reliably, and cost-effectively, in a decentralized setting.


Asunto(s)
Enfermedad Crónica/prevención & control , Inflamación/diagnóstico , Saliva/química , Biomarcadores/análisis , Biomarcadores/orina , Enfermedad Crónica/terapia , Humanos , Infecciones/diagnóstico , Inflamación/orina
19.
Arch Biochem Biophys ; 593: 80-9, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26874195

RESUMEN

Osteoporosis is a bone disease that is exacerbated by aging and age-associated chronic diseases such as cancer. Cancer-induced bone loss is usually treated with bisphosphonates or denosumab, an antibody against receptor activator of nuclear factor (NF)-κB ligand (RANKL). Because these drugs are expensive and have numerous side effects and high rates of toxicity, safer, more effective, and more affordable therapies for osteoporosis are still needed. We identified a compound, calebin A (CA), derived from turmeric (Curcuma longa) that affects osteoclastogenesis through modulation of the RANKL signalling pathway. The CA's effect on NF-κB activation was examined by electrophoretic mobility shift assay. Using mouse macrophages in vitro model, we found that CA suppressed RANKL-induced osteoclast differentiation of macrophages into osteoclasts, and downregulate RANKL-induced osteoclastogenesis-related marker gene expression, including NFATc-1, TRAP, CTR, and cathepsin K. CA also suppressed the osteoclastogenesis induced by multiple myeloma and breast cancer cells. This effect of CA was correlated with suppression of the phosphorylation and degradation of inhibitor of κB and, thus, inhibition of NF-κB activation. Furthermore, we found that an NF-κB-specific inhibitory peptide blocked RANKL-induced osteoclastogenesis, demonstrating that the NF-κB signalling pathway is mandatory for RANKL-induced osteoclastogenesis. Our results conclusively indicate that CA downmodulates the osteoclastogenesis induced by RANKL and by tumour cells through suppression of NF-κB pathway.


Asunto(s)
Cinamatos/farmacología , Curcuma/química , Monoterpenos/farmacología , Osteoclastos/efectos de los fármacos , Ligando RANK/antagonistas & inhibidores , Animales , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Humanos , Quinasa I-kappa B/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Fosforilación , Ligando RANK/metabolismo , Transducción de Señal
20.
Oncotarget ; 7(11): 13182-96, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26909608

RESUMEN

The development of chemoresistance in human pancreatic cancer is one reason for the poor survival rate for patients with this cancer. Because multiple gene products are linked with chemoresistance, we investigated the ability of ursolic acid (UA) to sensitize pancreatic cancer cells to gemcitabine, a standard drug used for the treatment of pancreatic cancer. These investigations were done in AsPC-1, MIA PaCa-2, and Panc-28 cells and in nude mice orthotopically implanted with Panc-28 cells. In vitro, UA inhibited proliferation, induced apoptosis, suppressed NF-κB activation and its regulated proliferative, metastatic, and angiogenic proteins. UA (20 µM) also enhanced gemcitabine (200 nM)-induced apoptosis and suppressed the expression of NF-κB-regulated proteins. In the nude mouse model, oral administration of UA (250 mg/kg) suppressed tumor growth and enhanced the effect of gemcitabine (25 mg/kg). Furthermore, the combination of UA and gemcitabine suppressed the metastasis of cancer cells to distant organs such as liver and spleen. Immunohistochemical analysis showed that biomarkers of proliferation (Ki-67) and microvessel density (CD31) were suppressed by the combination of UA and gemcitabine. UA inhibited the activation of NF-κB and STAT3 and the expression of tumorigenic proteins regulated by these inflammatory transcription factors in tumor tissue. Furthermore, the combination of two agents decreased the expression of miR-29a, closely linked with tumorigenesis, in the tumor tissue. UA was found to be bioavailable in animal serum and tumor tissue. These results suggest that UA can inhibit the growth of human pancreatic tumors and sensitize them to gemcitabine by suppressing inflammatory biomarkers linked to proliferation, invasion, angiogenesis, and metastasis.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/patología , Triterpenos/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Inflamación/patología , Ratones , Ratones Desnudos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina , Ácido Ursólico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...