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

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Mol Med ; 26(23): 5807-5819, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36308422

RESUMO

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide. Although cisplatin-based chemotherapy is commonly used in HNSCC, frequent development of cisplatin resistance is a potential cause of poor HNSCC prognosis. In the present study, we investigated the anticancer efficacy of a major paclitaxel metabolite namely 7-Epitaxol in cisplatin-resistant HNSCC. The findings revealed that 7-Epitaxol exerts cytotoxic effects in cisplatin-resistant HNSCC cell lines by inducing cell cycle arrest and intrinsic and extrinsic apoptotic pathways. Specifically, 7-Epitaxol increased Fas, TNF-R1, DR5, DcR3 and DcR2 expressions, reduced Bcl-2 and Bcl-XL (anti-apoptotic proteins) expressions, and increased Bid and Bim L/S (pre-apoptotic proteins) expressions, leading to activation of caspase-mediated cancer cell apoptosis. At the upstream cell signalling level, 7-Epitaxol reduced the phosphorylation of AKT, ERK1/2 and p38 to trigger apoptosis. In vivo results showed that animals treated with 7-Epitaxol show antitumor growth compared to control animals. Taken together, the study demonstrates the potential anticancer efficacy of 7-Epitaxol in inducing apoptosis of cisplatin-resistant HNSCC cells through the suppression of AKT and MAPK signalling pathways.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Animais , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Proteínas Proto-Oncogênicas c-akt , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Apoptose , Proteínas Reguladoras de Apoptose
2.
Molecules ; 27(10)2022 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-35630709

RESUMO

Foods rich in antioxidants such as lycopene have a major role in maintaining cardiac health. Lycopene, 80% of which can be obtained by consuming a common vegetable such as tomato, can prevent the disturbances that contribute to cardiovascular disease (CVD). The present work begins with a brief introduction to CVD and lycopene and its various properties such as bioavailability, pharmacokinetics, etc. In this review, the potential cardio-protective effects of lycopene that reduce the progression of CVD and thrombotic complications are detailed. Further, the protective effects of lycopene including in vitro, in vivo and clinical trials conducted on lycopene for CVD protective effects are explained. Finally, the controversial aspect of lycopene as a protective agent against CVD and toxicity are also mentioned.


Assuntos
Doenças Cardiovasculares , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/prevenção & controle , Carotenoides/farmacologia , Carotenoides/uso terapêutico , Fatores de Risco de Doenças Cardíacas , Humanos , Licopeno/uso terapêutico , Fatores de Risco
3.
Environ Toxicol ; 36(9): 1848-1856, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34076342

RESUMO

Head and neck cancer is associated with poor prognosis because of its highly metastatic nature. For the better management of head and neck cancer patients, it is very important to diagnose the cancer at an early stage, as well as to prevent the rapid spread of cancer either through direct invasion or lymphatic metastasis. In present study, the effect of dehydrocrenatidine, which is a beta-carboline alkaloid found in the medicinal plant Picrasma quassioides, on human head and neck cancer metastasis was investigated. The study results revealed the treatment of FaDu, SCC9, and SCC47 cells with 5, 10, and 20 µM of dehydrocrenatidine significantly decreased the motility, migration, and invasion of head and neck cancer cells. Moreover, the dehydrocrenatidine treatment significantly decreased the expression of MMP-2 and phosphorylation of ERK1/2 and JNK1/2. Additional experiments revealed that the cotreatment of dehydrocrenatidine with either ERK1/2 or JNK1/2 inhibitor caused further reduction in cancer cell motility and migration compared to that in dehydrocrenatidine treatment alone. Moreover, similar trend was observed in case of ERK1/2 and JNK1/2 phosphorylation and MMP-2 expression after the cotreatment. Taken together, the mechanism by which dehydrocrenatidine can decrease the phosphorylation of ERK1/2 and JNK1/2, follow decrease the expression of MMP-2 and inhibits head and neck cancer cells invasion and migration. This present study identifies dehydrocrenatidine as a potent antimetastatic agent that can be used clinically to improve head and neck cancer prognosis.


Assuntos
Neoplasias de Cabeça e Pescoço , Metaloproteinase 2 da Matriz , Carbolinas , Linhagem Celular Tumoral , Movimento Celular , Humanos , Sistema de Sinalização das MAP Quinases , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Invasividade Neoplásica
4.
Crit Rev Food Sci Nutr ; 60(20): 3522-3546, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31822111

RESUMO

Most of the previous studies in last three decades report evidence of interactions between the different phytochemicals and the proteins involved in signal transduction pathways using in silico, in vitro, ex vivo, and in vivo analyses. However, extrapolation of these findings for clinical purposes has not been that fruitful. The efficacy of the phytochemicals in vivo studies is limited by parameters such as solubility, metabolic degradation, excretion, etc. Various approaches have now been devised to circumvent these limitations. Recently, chemical modification of the phytochemicals are demonstrated to reduce some of the limitations and improve their efficacy. Similar to traditional medicines several combinatorial phytochemical formulations have shown to be more efficient. Further, phytochemicals have been reported to be even more efficient in the form of nanoparticles. However, systematic evaluation of their efficacy, mode of action in pathway modulation, usage and associated challenges is required to be done. The present review begins with basic understanding of how signaling cascades regulate cellular response and the consequences of their dysregulation further summarizing the developments and problems associated with the dietary phytochemicals and also discuss recent approaches in strengthening these compounds in pharmacological applications. Only context relevant studies have been reviewed. Considering the limitations and scope of the article, authors do not claim inclusion of all the early and recent studies.


Assuntos
Nanopartículas , Compostos Fitoquímicos , Frutas , Compostos Fitoquímicos/farmacologia , Transdução de Sinais
5.
J Cell Physiol ; 234(9): 16125-16135, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30741420

RESUMO

In this present study, Oreochromis mossambicus tilapia were transferred to cold water at 12°C for various time intervals (1, 4, 8, 24, and 48 hr) and its innate immune response was analyzed by studying cellular and humoral parameters. In vivo, alternative complement pathway activity in blood plasma was rapidly increased at 1 hr of cold water (12°C) exposure. Lysozyme activity and cortisol levels of plasma were increased at 4 and 1 hr, respectively. Surprisingly, only plasma cortisol levels remained unchanged through 24 hr of cold water transfer. Phagocytic ability, phagocytic capacity, and respiratory burst (RB) activity of head kidney (HK) leukocytes and splenocytes showed no any significant changes. In peripheral blood leukocytes, phagocytic capacity, and RB activity were increased at 24 hr of cold water exposure. The expressions of genes involved innate immunity in splenocytes and HK leukocytes of tilapia cold water exposure were analyzed, messenger RNA (mRNA) expressions of HSP70, HSP90, and immunoglobulin M failed to change upon exposure to cold stress. Major histocompatibility complex-I and II mRNAs were significantly increased in tilapia splenocytes at 1 hr of cold water transferred. Whereas myxovirus (Mx) expression was increased in splenocytes and HK leukocytes of tilapia after 1 hr of cold water exposed. Our result reveals that the exposure of tilapia to acute cold stress condition significantly enhances plasma acid phosphatase activity and both phagocytic capacity and RB activity. Furthermore, cold stress significantly stimulates Mx gene expression in splenocytes and HK leukocytes.

6.
J Cell Physiol ; 234(6): 8443-8454, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30565686

RESUMO

Engineering/reprogramming differentiated adult somatic cells to gain the ability to differentiate into any type of cell lineage are called as induced pluripotent stem cells (iPSCs). Offering unlimited self-renewal and differentiation potential, these iPSC are aspired to meet the growing demands in the field of regenerative medicine, tissue engineering, disease modeling, nanotechnology, and drug discovery. Biomaterial fabrication with the rapid evolution of technology increased their versatility and utility in regenerative medicine and tissue engineering, revolutionizing the stem cell biology research with the property to guide the process of proliferation, differentiation, and morphogenesis. Combining traditional culture platforms of iPSC with biomaterials aids to overcome the limitations associated with derivation, proliferation, and maturation, thereby could improve the clinical translation of iPSC. The present review discusses in brief about the reprogramming techniques for the derivation iPSC and details on several biomaterial guided differentiation of iPSC to different cell types with specific relevance to tissue engineering/regenerative medicine.


Assuntos
Materiais Biocompatíveis/farmacologia , Diferenciação Celular/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/citologia , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Autorrenovação Celular/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Medicina Regenerativa
7.
J Gene Med ; 21(12): e3142, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31721380

RESUMO

BACKGROUND: Cigarette smoking in women is raising a public health problem. The X-linked monoamine oxidase A (MAOA) was considered as a susceptibility gene to substance abuse of tobacco, but the evolutionary effect of MAOA may lead to a positive or negative association between genetic variations and smoking development among study regions. METHODS: Based on linkage disequilibrium (LD), we performed a haplotype-based association to explore the effect of MAOA gene on women's smoking risk in a case-control study. RESULTS: Genotyped single nucleotide polymorphisms (SNPs) of MAOA gene, rs5953210G>A, rs2283725A>G and rs1137070T>C, were significantly associated with current smoking risk in women, and the increased level of plasma MAO-A activity was raised with per copy increment of risk allele in current smokers (P < .01). The haplotype patterns with minor haplotype frequency >.05 were constructed using the Expectation-Maximization algorithm, and the haplotype-specific A-G-C pattern raised the 2-fold risk to develop regular smoking (P = .0005). In the diplotype analysis based on X-inactivation mechanism relative to no and full dosage compensation, we showed that A-G-C haplotype not only increased regular smoking risk in a dose-dependent manner (Ptrend = .0011) but also contributed to smoking risk in the dosage compensation mechanism. Compared to non-smokers, the effect of A-G-C haplotype on random X-activation was associated with the raised MAO-A activity in women smokers (P < .05) although the lifetime cigarette consumption showed a difference that was not statistically significant. CONCLUSION: This study provides information on MAOA LD-based haplotype and diplotype patterns in women smoking.


Assuntos
Alelos , Haplótipos , Desequilíbrio de Ligação , Monoaminoxidase/genética , Fumar/genética , Adulto , Fatores Etários , Idoso , Ativação Enzimática , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Humanos , Pessoa de Meia-Idade , Monoaminoxidase/metabolismo , Razão de Chances , Polimorfismo de Nucleotídeo Único
8.
Environ Toxicol ; 34(12): 1320-1328, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31486215

RESUMO

Carthamus tinctorius L. (Compositae) is used in Chinese medicine to treat heart disease and inflammation. In our previous study, we found that C. tinctorius L. inhibited lipopolysaccharides (LPS)-induced tumor necrosis factor-alpha (TNF-α) activation, JNK expression, and apoptosis in H9c2 cardiomyoblast cells. The present study was performed to investigate the protective effect of C. tinctorius extract (CTF) on LPS-challenged H9c2 myocardioblast cell and to explore the possible underlying mechanism. Cell viability assay showed that LPS treatment decreased the cell viability of H9c2 cell, whereas CTF treatment reversed LPS cytotoxicity in a dose-dependent manner, especially in the LPS + CTF 25 (µg/mL) group. LPS treatment-induced apoptosis was determined by transferase-mediated dUTP nick end labeling assay, and by Western blot. LPS-induced apoptotic bodies were decreased following CTF treatment. Expression of TNF-α, FAS-L, FAS, FADD, caspase-8, BID, and t-BID was significantly increased in LPS-treated H9c2 cells. In contrast, it was significantly suppressed by the administration of CTF extract. In addition, CTF treatment activates antiapoptotic proteins, Bcl-2 and p-Bad, and downregulates Bax, cytochrome-c, caspase-9, caspase-3, and apoptosis-inducing factor expression. Furthermore, CTF exerted cytoprotective effects by activating insulin-like growth factor-I (IGF-I) signaling pathway leading to downregulation of the apoptotic proteins involved in FAS death receptor pathway. In addition, AG1024 and IGF-I receptor (IGF-IR) inhibitor and siRNA silencing reverses the effect of CTF implying that CTF functions through the IGF-IR pathway to inhibit LPS-induced H9c2 apoptosis. These results suggest that treatment with CTF extract prevented the LPS-induced apoptotic response through IGF-I pathway.


Assuntos
Apoptose/efeitos dos fármacos , Carthamus tinctorius/química , Extratos Vegetais/farmacologia , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor fas/metabolismo , Animais , Carthamus tinctorius/metabolismo , Caspase 3/metabolismo , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Extratos Vegetais/química , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Fator de Necrose Tumoral alfa/metabolismo
9.
J Cell Physiol ; 233(11): 8458-8466, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29856483

RESUMO

Heart failure (HF) remains a major cause of morbidity and mortality worldwide. The primary cause identified for HF is impaired left ventricular myocardial function, and clinical manifestations may lead to severe conditions like pulmonary congestion, splanchnic congestion, and peripheral edema. Development of new therapeutic strategies remains the need of the hour for controlling the problem of HF worldwide. Deeper insights into the molecular mechanisms involved in etiopathology of HF indicate the significant role of calcium signaling, autocrine signaling pathways, and insulin-like growth factor-1 signaling that regulates the physiologic functions of heart growth and development such as contraction, metabolism, hypertrophy, cytokine signaling, and apoptosis. In view of these facts, a transcription factor (TF) regulating the myriad of these signaling pathways may prove as a lead candidate for development of therapeutics. Adenovirus E4 promoter-binding protein (E4BP4), also known as nuclear-factor, interleukin 3 regulated (NFIL3), a type of basic leucine zipper TF, is known to regulate the signaling processes involved in the functioning of heart. The current review discusses about the expression, structure, and functional role of E4BP4 in signaling processes with emphasis on calcium signaling mechanisms, autocrine signaling, and insulin-like growth factor II receptor-mediated processes regulated by E4BP4 that may regulate the pathogenesis of HF. We propose that E4BP4, being the critical component for the regulation of the above signaling processes, may serve as a novel therapeutic target for HF, and scientific investigations are merited in this direction.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Proteínas de Ligação a DNA/genética , Insuficiência Cardíaca/genética , Apoptose/genética , Comunicação Autócrina/genética , Sinalização do Cálcio/genética , Regulação da Expressão Gênica/genética , Insuficiência Cardíaca/patologia , Humanos , Fator de Crescimento Insulin-Like I/genética , Regiões Promotoras Genéticas , Fatores de Transcrição/genética
10.
Environ Toxicol ; 33(2): 234-247, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29134746

RESUMO

The physiological regulation of Oestrogen receptor α (ERα) and peroxisome proliferator-activated receptor alpha (PPARα) in Hepatocellular carcinoma (HCC) remains unknown. The present study we first treat the cells with fenofibrate and further investigated the possible mechanisms of 17ß-estradiol (E2 ) and/or ERα on regulating PPARα expression. We also found higher PPARα expression in the tumor area than adjacent areas and subsequently compared PPARα expression in four different hepatic cancer cell lines. Hep3B cells were found to express more PPARα than the other cell lines. Using the PPARα agonist fenofibrate, we found that fenofibrate increased Hep3B cell proliferation efficiency by increasing cell cycle proteins, such as cyclin D1 and PCNA, and inhibiting p27 and caspase 3 expressions. Next, we performed transient transfections and immuno-precipitation studies using the pTRE2/ERα plasmid to evaluate the interaction between ERα and PPARα. ERα interacted directly with PPARα and negatively regulated its function. Moreover, in Tet-on ERα over-expressed Hep3B cells, E2 treatment inhibited PPARα, its downstream gene acyl-CoA oxidase (ACO), cyclin D1 and PCNA expression and further increased p27 and caspase 3 expressions. However, over-expressed ERα plus 17-ß-estradiol (10-8 M) reversed the fenofibrate effect and induced apoptosis, which was blocked in ICI/melatonin/fenofibrate-treated cells. This study illustrates that PPARα expression and function were negatively regulated by ERα expression in Hep3B cells.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Fenofibrato/toxicidade , Hipolipemiantes/toxicidade , PPAR alfa/metabolismo , Regulação para Cima/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , PPAR alfa/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ligação Proteica , RNA Mensageiro/metabolismo
11.
Molecules ; 23(8)2018 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-30082655

RESUMO

In this study, we investigated whether 16-hydroxycleroda-3,13-dien-15,16-olide (HCD) and N-methyl-actinodaphine (MA) could sensitize breast cancer cells to Tamoxifen (TMX) treatment. MA or HCD alone or in combination with TMX dose-dependently inhibited MCF-7 and MDA-MB-231 cell growth, with a more potent inhibition on MDA-MB 231 cells. Furthermore, this novel combination significantly induced S and G2/M cell cycle phase in MDA-MB 231 than MCF-7 cells. Further determination of the apoptotic induction showed that MA or HCD and TMX combination inhibited MDA-MB-231 and MCF-7 cancer cells by upregulating Bax and by downregulating Bcl-2 mRNA and protein expression without altering Caspase-8 and Caspase-12 expression. These results suggest that MA or HCD pretreatment may potentiate the anti-tumor effect of tamoxifen on breast cancer.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Dioxolanos/farmacologia , Tamoxifeno/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Caspase 12/metabolismo , Caspase 8/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dioxolanos/química , Feminino , Humanos , Células MCF-7
12.
Molecules ; 23(10)2018 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-30262792

RESUMO

Neurodegenerative diseases are normally distinguished as disorders with loss of neurons. Various compounds are being tested to treat neurodegenerative diseases (NDs) but they possess solitary symptomatic advantages with numerous side effects. Accumulative studies have been conducted to validate the benefit of phytochemicals to treat neurodegenerative diseases including Alzheimer's disease (AD) and Parkinson's disease (PD). In this present review we explored the potential efficacy of phytochemicals such as epigallocatechin-3-galate, berberin, curcumin, resveratrol, quercetin and limonoids against the most common NDs, including Alzheimer's disease (AD) and Parkinson's disease (PD). The beneficial potentials of these phytochemicals have been demonstrated by evidence-based but more extensive investigation needs to be conducted for reducing the progression of AD and PD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Fármacos Neuroprotetores , Doença de Parkinson/tratamento farmacológico , Compostos Fitoquímicos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Humanos , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Compostos Fitoquímicos/química , Compostos Fitoquímicos/uso terapêutico
13.
J Cell Physiol ; 232(11): 3020-3029, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28500736

RESUMO

In our previous study palmitic acid (PA) induced lipotoxicity and switches energy metabolism from CD36 to GLUT4 in H9c2 cells. Low level of high density lipoprotein (HDL) is an independent risk factor for cardiac hypertrophy. Therefore, we in the present study investigated whether HDL can reverse PA induced lipotoxicity in H9c2 cardiomyoblast cells. In this study, we treated H9c2 cells with PA to create a hyperlipidemia model in vitro and analyzed for CD36 and GLUT4 metabolic pathway proteins. CD36 metabolic pathway proteins (phospho-AMPK, SIRT1, PGC1α, PPARα, CPT1ß, and CD36) were decreased by high PA (150 and 200 µg/µl) concentration. Interestingly, expression of GLUT4 metabolic pathway proteins (p-PI3K and pAKT) were increased at low concentration (50 µg/µl) and decreased at high PA concentration. Whereas, phospho-PKCζ, GLUT4 and PDH proteins expression was increased in a dose dependent manner. PA treated H9c2 cells were treated with HDL and analyzed for cell viability. Results showed that HDL treatment induced cell proliferation efficiency in PA treated cells. In addition, HDL reversed the metabolic effects of PA: CD36 translocation was increased and reduced GLUT4 translocation, but HDL treatment significantly increased CD36 metabolic pathway proteins and reduced GLUT4 pathway proteins. Rat neonatal cardiomyocytes showed similar results. In conclusion, HDL reversed palmatic acid-induced lipotoxicity and energy metabolism imbalance in H9c2 cardiomyoblast cells and in neonatal rat cardiomyocyte cells.


Assuntos
Antígenos CD36/metabolismo , Metabolismo Energético/efeitos dos fármacos , Transportador de Glucose Tipo 4/metabolismo , Lipoproteínas HDL/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Ácido Palmítico/toxicidade , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Animais Recém-Nascidos , Cardiotoxicidade , Carnitina O-Palmitoiltransferase/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Citoproteção , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , PPAR alfa/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação , Transporte Proteico , Proteólise , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Sirtuína 1/metabolismo , Fatores de Tempo
14.
Mol Cell Biochem ; 434(1-2): 181-195, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28577190

RESUMO

The present study was aimed to investigate the protective effects of 17ß-estradiol (E2) and estrogen receptor α (ERα) on isoproterenol (ISO)-treated H9c2 cardiomyoblast cells. In the present study, we treated H9c2 cells with ISO, a ß-adrenergic receptor agonist, to induce myocardiac hypertrophy. Pre-administration of E2 or ERα (induced by doxycycline) and E2 plus ERα significantly prevented ISO-induced increase of cell size and cytosolic calcium accumulation, accompanied with increased mRNA of atrial natriuretic peptide and brain natriuretic peptide. However, ICI-ERs antagonist, and melatonin, a specific inhibitor for ERα, reversed the cardioprotective effects, suggesting that E2 action was mediated through ERα. Further evidences showed that E2 and ERα increased the protein level of GSK3ß and protein phosphatase 2a inhibitor 2 (I2-PP2A), which subsequently enhanced the activation of I2-PP2A by disrupting PP2A activity and maintains normal calcium outflow. Collectively, E2 and ERα inhibited hypertrophy by preventing cytosol calcium accumulation and by inhibiting the association between PP2A with Na+-Ca2+ exchanger via GSK3ß and I2-PP2A activation.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Fator Natriurético Atrial/metabolismo , Cálcio/metabolismo , Estradiol/fisiologia , Receptor alfa de Estrogênio/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Isoproterenol/farmacologia , Fatores de Transcrição NFATC/metabolismo , Proteína Fosfatase 2/metabolismo , Animais , Linhagem Celular
15.
Environ Toxicol ; 32(1): 290-301, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26790859

RESUMO

Marine organisms are proven to be rich source of secondary metabolites that can be used to treat various diseases. Excavatolide B (Exc.B), the most abundant metabolite was found in the marine coral Briareum excavatum exhibits cytotoxic effects against lung cancer cell. Treatment of the A549 cells with Exc.B significantly reduced its cell viability and induced cell cycle arrest at subG1 phase in a dose- and time-dependent manner, respectively. Apoptosis induction by Exc.B was further confirmed by decreased pro-caspase 3 expressions and increased proteolytic cleavage of poly (ADP-ribose) polymerase (PARP) expression. Furthermore, Exc.B increased reactive oxygen species (ROS) and reactive nitrogen species (RNS) and also decreased the antioxidant enzymes such as, Catalase, GPx, SOD, GST, and GSH. The proteomic analysis data revealed that total thirty six proteins were altered by Exc.B. STRING database showed that most of the altered proteins have no interaction between each other. Based on these data, KSR1, RuVBL2, PPAR-γ, and Tenascin X proteins were chosen to validate the 2DE data by Western blotting. Additional experiments demonstrated that Exc.B induced PTEN expression and inhibited pAKT and NF-kB expression. These results provide a novel insight into mechanisms underlying the inhibition of A549 cells growth by excavatolide B. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 290-301, 2017.


Assuntos
Proliferação de Células/efeitos dos fármacos , Diterpenos/toxicidade , Expressão Gênica/efeitos dos fármacos , PPAR gama/metabolismo , Células A549 , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Caspase 3/metabolismo , Catalase/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Eletroforese em Gel Bidimensional , Glutationa Peroxidase/metabolismo , Glutationa Transferase/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , NF-kappa B/genética , NF-kappa B/metabolismo , PPAR gama/genética , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Espectrometria de Massas em Tandem
16.
Environ Toxicol ; 32(2): 519-529, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26990689

RESUMO

In our previous experiments, we found ß-catenin was highly expressed in the tumor area with high invasive ability and poor prognosis. In this study, we have examined the mechanism by which ERα regulates ß-catenin expression as well as the metastasis ability of hepatocellular cancer HA22T cells. To identify whether the anticancer effect of estrogen and ERα is mediated through suppression of ß-catenin expression, we co-transfected pCMV-ß-catenin and ERα into HA22T cells, and determined the cell motility by wound healing, invasion, and migration assays. Results showed that estrogen and/or ERα inhibited ß-catenin gene expression and repressed HA22T cell motility demonstrated that similar data was observed in cells expressing the ERα stable clone. Moreover, we examined the protein-protein interaction between ERα and ß-catenin by immunostain, co-immunoprecipitation, and Western blotting. E2 enhanced the binding of ERα with ß-catenin and then triggered ß-catenin to bind with E3 ligase (ßTrCP) to promote ß-catenin degradation. Finally by employing systematic ChIP studies, we showed ERα can interact directly with the ß-catenin promoter region following E2 treatment. All our results reveal that estrogen and ERα blocked metastatic function of HA22T cells by modulating GSK3ß and ßTrCP expression and further enhanced ß-catenin degradation and suppressed its downstream target genes. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 519-529, 2017.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , beta Catenina/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular , Regulação para Baixo , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Imuno-Histoquímica , Imunoprecipitação , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Microscopia de Fluorescência , Proteínas Contendo Repetições de beta-Transducina/genética
17.
Environ Toxicol ; 31(12): 2045-2056, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26464283

RESUMO

Helioxanthin, an active compound from Taiwania cryptomerioides Hayata, has been shown to have various biological activities. However, their anticancer effect in oral squamous cell carcinoma has not been well established yet. Helioxanthin inhibited the proliferation of oral squamous cell carcinoma cells in a dose-dependent manner by inducing G2/M phase arrest. Similarly, helioxanthin inhibited cyclooxygenase-2, (COX-2), phosphorylated EGFR, and extracellular-signal-regulated kinases (ERK) protein level and further reduced the nuclear accumulation of phosphorylated epidermal growth factor receptor (pEGFR) and activator protein-1(AP-1) family protein, c-fos. Moreover, helioxanthin at the dose of 20 and 30 mg kg-1 for 15 days reduced the tumor growth in animal model. This study demonstrated that Helioxanthin exerts its anticancer activity against oral cancer cells by downregulating EGFR/ERK/c-fos signaling pathway to inhibit COX-2 level and by activating cyclin-dependent kinase inhibitor (p27) to further induce G2/M cell cycle arrest. This helioxanthin may serve as a novel candidate for oral cancer prevention. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 2045-2056, 2016.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Lignanas/farmacologia , Neoplasias Bucais/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Arecolina , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Xenoenxertos , Lignanas/uso terapêutico , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos Nus , Neoplasias Bucais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo
18.
Tzu Chi Med J ; 35(3): 242-246, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37545799

RESUMO

Objectives: This study retrospectively evaluated the effectiveness of percutaneous pulley release by our newly designed needle knife in terms of cure, relapse, and complication rates. Materials and Methods: Two hundred and fifty-seven patients were allocated into male and female groups between October 2014 and September 2021. We included patients >15 years of age with a trigger finger (TF) (types II-VI). The primary outcome was the absence of a TF and pain-free movement. In contrast, the secondary outcome included second-time surgery and the number of complications such as infection and admission for antibiotics. Results: One hundred patients were male, and 157 patients were female. Males and females had mean ages of 62.45 ± 11.76 and 61.50 ± 8.57 years, respectively. The operative time was significantly longer in males than in females (7.88 ± 6.02 vs. 6.52 ± 3.74 min in males and females, respectively, P = 0.027). However, the percentages of diabetes mellitus and gout were the same in both groups. For the percutaneous methods with our needle knife, remission of the trigger was achieved in all cases. In addition, seven patients received revision and three patients with complications. After needle surgery, topical and joint pain scores were improved in both groups (from 5.09 ± 1.31 to 0.80 ± 1.56). Conclusion: The percutaneous methods with our needle knife displayed effectiveness. The cure rate was high, and the relapse rate was low. Further large-scale clinical trials comparing percutaneous needle to open surgery for releasing the TF will be needed to confirm our results.

19.
Pharmaceuticals (Basel) ; 15(4)2022 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-35455398

RESUMO

Liver cancer is a leading cause of death worldwide. Despite advancement in therapeutic interventions, liver cancer is associated with poor prognosis because of highly lethal characteristics and high recurrence rate. In the present study, the anticancer potential of a plant-based alkaloid namely dehydrocrenatidine has been evaluated in human liver cancer cells. The study findings revealed that dehydrocrenatidine reduced cancer cell viability by arresting cell cycle at G2/M phase and activating mitochondria-mediated and death receptor-mediated apoptotic pathways. Specifically, dehydrocrenatidine significantly increased the expression of extrinsic pathway components (FAS, DR5, FADD, and TRADD) as well as intrinsic pathway components (Bax and Bim L/S) in liver cancer cells. In addition, dehydrocrenatidine significantly increased the cleavage and activation of PARP and caspases 3, 8, and 9. The analysis of upstream signaling pathways revealed that dehydrocrenatidine induced caspase-mediated apoptosis by suppressing the phosphorylation of JNK1/2. Taken together, the study identifies dehydrocrenatidine as a potent anticancer agent that can be use clinically to inhibit the proliferation of human liver cancer cells.

20.
J Food Biochem ; 46(5): e14041, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35064587

RESUMO

Growing evidences indicate that high glucose toxicity-associated fibrotic effects play a pivotal role in diabetic nephropathy (DN). Tubular epithelial-myofibroblast transdifferentiation is a major hallmark of renal fibrosis event under diabetic stress. Roots of Glycyrrhiza uralensis (Radix glycyrrhizae) used as a sweetener and traditional Chinese medicine possess high potential for renal protection. In this study, a cell model for high glucose (HG) injury with HK-2 renal proximal tubular epithelial cell line and a type-II-diabetes model with Apoeem1/Narl /Narl mice was established and the beneficial effects of aqueous R. glycyrrhizae extract (RGE) was investigated. RGE-induced regulation on the high glucose-induced excessive production of TGF-ß1 and the Smad/Stat3 mechanisms of renal fibrosis were determined. HK-2 cells were challenged with 45 mM of high glucose for 48 hr. Following high glucose challenge, the cells were treated with 0.5, 1, and 1.5 mg/ml concentrations of RGE. The effect of RGE on DN was determined using high fructose diet-induced type-II-diabetes in Apoeem1/Narl /Narl mice models. Our results showed that RGE suppressed the expression of HG-induced TGFß signaling and associated fibrosis mechanism better than the pharmacological drug acarbose. These data suggest that RGE as a potential herbal supplement in attenuating fibrosis-associated diabetic nephropathy and a potential agent in diabetes treatments.


Assuntos
Nefropatias Diabéticas , Medicamentos de Ervas Chinesas , Glycyrrhiza uralensis , Animais , Linhagem Celular , Transdiferenciação Celular , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Fibrose , Glucose , Glycyrrhiza uralensis/química , Humanos , Camundongos , Miofibroblastos/metabolismo , Extratos Vegetais/farmacologia , Transdução de Sinais , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA