Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Environ Toxicol ; 39(2): 840-856, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37853854

RESUMO

INTRODUCTION: Hepatocellular carcinoma (HCC) is a common solid cancer and the leading cause of cancer deaths worldwide. Sorafenib is the first drug used to treat HCC but its effectiveness needs to be improved, and it is important to find ways to treat cancer that combine sorafenib with other drugs. Synergistic therapies lower effective drug doses and side effects while enhancing the anticancer effect. PURPOSE: In the present study, the therapeutic potential of sorafenib in combination with escin and its underlying mechanism in targeting liver cancer has been established. STUDY DESIGN/METHODS: The IC50 of sorafenib and escin against HepG2, PLC/PRF5 and Huh7 cell lines were determined using MTT assay. The combination index, dose reduction index, isobologram and concentrations producing synergy were evaluated using the Chou-Talaly algorithm. The sub-effective concentration of sorafenib and escin was selected to analyze cytotoxic synergistic potential. Cellular ROS, mitochondrial membrane potential, annexin V and cell cycle were evaluated using a flow-cytometer, and autophagy biomarkers were determined using western blotting. Moreover, autophagy was knocked down using ATG5 siRNA to confirm its role. A DEN-induced liver cancer rat model was developed to check the synergy of sorafenib and escin. RESULTS: Different concentrations of escin reduced the IC50 of sorafenib in HepG2, PLC/PRF5 and Huh7 cell lines. Chou-Talaly algorithm determined cytotoxic synergistic concentrations of sorafenib and escin in these cell lines. Mechanistically, this combination over-expressed p62 and LC-II, reflecting autophagy block and induced late apoptosis, further reconfirmed by ATG5 knockdown. Sorafenib and escin combination  reduced HCC serum biomarker α-feto protein (α-FP) by 1.5 folds. This combination restricted liver weight, tumor number and size, also, conserved morphological features of liver cells. The combination selectively targeted the G0 /G1 phase of cancer cells. CONCLUSION: Escin and sorafenib combination potentially up-regulates p62 to block autophagy to induce late apoptosis in liver cancer cells.


Assuntos
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Ratos , Antineoplásicos/farmacologia , Apoptose , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células , Escina/farmacologia , Neoplasias Hepáticas/patologia , Proteínas Associadas aos Microtúbulos , Sorafenibe/farmacologia
2.
Phytother Res ; 37(10): 4819-4837, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37468281

RESUMO

Combining anti-cancer drugs has been exploited as promising treatment strategy to target lung cancer. Synergistic chemotherapies increase anti-cancer effect and reduce effective drug doses and side effects. In this study, therapeutic potential of escin in combination with sorafenib has been explored. 3-(4,5-Dimethylthiazol-2-yl)-2 5-diphenyltetrazolium bromide assay was used to calculate IC50 values. The synergy was evaluated using Chou-Talaly algorithm. Cellular reactive oxygen species, mitochondrial membrane potential, annexin V, and cell-cycle studies were done by flow-cytometer, and autophagy biomarkers expression were determined using western blotting. Moreover, autophagy was knocked down using ATG5 siRNA to confirm its role, diethylnitrosamine-induced lung cancer model was used to check the synergy of sorafenib/escin. Escin significantly reduced the IC50 of sorafenib in A549 and NCIH460 cells. The combination of sorafenib/escin produced a 2.95 and 5.45 dose reduction index for sorafenib in A549 and NCI-H460 cells. The combination of over-expressed p62 and LC3-II reflects autophagy block-mediated late apoptosis. This phenomenon was reconfirmed by ATG5 knockdown. This combination also selectively targeted G0/G1 phase of cancer cells. In in vivo study, the combination reduced tumour load and lower elevated serum biochemical parameters. The combination of sorafenib/escin synergistically inhibits autophagy to induce late apoptosis in lung cancer cells' G0/G1 phase.

3.
Phytother Res ; 35(5): 2418-2428, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33254282

RESUMO

Purpurin is a naturally occurring anthraquinone isolated from the roots of Rubia cordifolia. Historically, it has been used as a red dye. However, its photosensitizing property and biological effects have deciphered its novel application. Purpurin shows antigenotoxic, anticancer, neuromodulatory, and antimicrobial potential associated with antioxidant action in in vivo and in vitro experiments. A robust antioxidant nature of purpurin is responsible for the majority of its pharmacological effects. It produces anti-inflammatory activity by reducing oxidative stress, which is a fundamental property to target diseases involving endoplasmic reticulum and mitochondrial stress. It can cross the blood-brain barrier and produce neuroprotective effects, including antidepressant and anti-Alzheimer action. It shows antimutagenic property via inhibiting essential CYP-450 enzymes. Interestingly, it gets photosensitized by UV-light and produces target-specific ROS-dependent apoptosis in cancer cells. Therefore, it owns cell killing and cell survival potential subject to the influence of external conditions. Hitherto, limited research studies are performed with purpurin to understand its therapeutic potential. Hence, this review describes and discusses different in vivo, in vitro, and in silico studies performed using purpurin. It also covers physicochemical, pharmacokinetics, and toxicology aspects of purpurin. Moreover, in the end, the prospect of purpurin in the management of cancer has also been proposed.

4.
Pharmacol Res ; 146: 104282, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31129179

RESUMO

Cancer is amongst the life-threatening public health issue worldwide, hence responsible for millions of death every year. It is affecting human health regardless of their gender, age, eating habits, and ecological location. Many drugs and therapies are available for its cure still the need for effective targeted drugs and therapies are of paramount importance. In the recent past, Ca2+ signalling (including channels/transporters/pumps) are being studied as a plausible target for combating the cancer menace. Many evidence has shown that the intracellular Ca2+ homeostasis is altered in cancer cells and the remodelling is linked with tumor instigation, angiogenesis, progression, and metastasis. Focusing on these altered Ca2+ signalling tool kit for cancer treatment is a cross-cutting and emerging area of research. In addition, there are numerous phytomolecules which can be exploited as a potential Ca2+ (channels/transporters/ pumps) modulators in the context of targeting Ca2+ signalling in the cancer cell. In the present review, a list of plant-based potential Ca2+ (channel/transporters/pumps) modulators has been reported which could have application in the framework of repurposing the potential drugs to target Ca2+ signalling pathways in cancer cells. This review also aims to gain attention in and support for prospective research in this field.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Neoplasias/metabolismo , Compostos Fitoquímicos/farmacologia , Animais , Canais de Cálcio/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Compostos Fitoquímicos/uso terapêutico
5.
Int J Biol Macromol ; 257(Pt 1): 127945, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37951434

RESUMO

MicroRNAs (miRNAs) play a crucial role in cancer progression by selectively inducing translational degradation of messenger RNA (mRNA) via sequence-specific interactions with the 3'-untranslated region (3'-UTR). The potential targeting of miRNA has been recognized as a significant avenue for investigating the biological progression of diverse cancer types. Consequently, targeting of pri-miRNA and pre-miRNA by phytochemicals emerges as a viable strategy in the realm of anticancer therapies. Among phytochemicals, triterpenoids have garnered significant recognition for their chemotherapeutic and chemopreventive capabilities in combating multiple cancers. To date, there is a dearth of literature about the molecular interactions between triterpenoids and miRNAs. The primary objective of this investigation is to discern the potential triterpenoids that can function as modulators for specific miRNAs, namely pri-miRNA-19b-2, pre-miR21, microRNA 20b, pri-miRNA-208a, pri-miRNA-378a, pri-miRNA-320b-2, and pri-miRNA-300, achieved through the use of in silico investigations. The study primarily focused on performing drug-likeness, computer-aided toxicity, and pharmacokinetic prediction studies for triterpenoids. Furthermore, molecular docking and simulation techniques were employed to investigate these compounds. The triterpenoids studied were shown to have drug-likeness characteristics, although asiatic acid, lupeol, and pristimerin were able to pass all toxicity tests. Among the triterpenoids that underwent docking, pristimerin had a significant binding energy of -10.9 kcal/mol during its interaction with pri-miR-378a. The stable interaction between the pristimerin and miRNA complex was demonstrated by molecular dynamics simulation. As a result, pristimerin has the potential to act as a modulator of carcinogenic miRNAs, making it a promising candidate for cancer prevention and treatment due to its tailored modulation of miRNA activity.


Assuntos
MicroRNAs , Neoplasias , Triterpenos Pentacíclicos , Triterpenos , Humanos , Processamento Pós-Transcricional do RNA , Triterpenos/farmacologia , Angiogênese , Simulação de Acoplamento Molecular , Precursores de RNA/metabolismo , MicroRNAs/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Proliferação de Células
6.
Toxicol In Vitro ; 93: 105666, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37611852

RESUMO

Combination therapy has been proposed as a promising approach for lung cancer treatment, as it can enhance anticancer efficacy, and reduce dosages and adverse effects. This study aimed to explore the therapeutic potential of gossypol, a natural polyphenolic compound with sorafenib for treating lung cancer cells and elucidating its mechanism of action. The MTT assay was utilized to determine the IC50 of sorafenib and gossypol against A549 and NCI H460 cell lines. The Chou-Talaly algorithm was employed to determine the combination index (CI). A sub-effective concentration of sorafenib and gossypol was chosen to investigate the possibility of cytotoxic synergy. Autophagy biomarkers were identified using Western blotting, and the function of autophagy was determined using ATG5 siRNA. Results show that IC50 of sorafenib significantly reduced in A549 and NCI H460 cells when co-treated with gossypol. The combination treatment showed a synergistic cytotoxic effect against tested cell lines. The Chou-Talaly algorithm confirmed sorafenib's dose reduction index (DRI) up to 3.86. In A549 cells, combination treatment down-regulated p62 and up-regulated LC3-II, indicating the initiation of autophagy-dependent cytotoxicity. This was further confirmed by siRNA ATG5 knockdown. Additionally, the combination treatment exclusively targeted G0/G1 phase cancer cells. In conclusion, the combination of gossypol and sorafenib shows a synergistic increase in the cytotoxic effect by promoting autophagy and apoptosis.


Assuntos
Antineoplásicos , Gossipol , Neoplasias Pulmonares , Humanos , Sorafenibe/farmacologia , Gossipol/farmacologia , Linhagem Celular Tumoral , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Antineoplásicos/farmacologia , Apoptose , Autofagia , RNA Interferente Pequeno/farmacologia , Proliferação de Células
7.
Int J Biol Macromol ; 252: 126328, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37579900

RESUMO

Lung cancer is the most common and lethal cancer worldwide, yet there are no adequate and novel medications to control this illness. Previous reports suggested the potential of protein kinases to target lung cancer by regulating autophagy. This study establishes the role of aescin, a triterpenoid saponin, in targeting protein kinases responsible for lung cancer proliferation and mobility. The experimental data revealed that aescin significantly impedes lung cancer cell proliferation by downregulating protein kinases such as AKT, mTOR, MEK, and ERK. Downregulation of AKT-mTOR may promote a string of events inducing cytotoxic autophagy-mediated apoptosis in the presence of aescin. Besides, aescin decreases mobility and invasion by downregulating HIF-1α and VEGF gene expressions. Moreover, it successfully monitors EGFR gene expression, improves lung histology, and regulates biochemical parameters in a pre-clinical DEN-induced lung cancer model. Aescin was observed to be safe and non-toxic in both in silico toxicity predictions and ex vivo erythrocyte fragility assays. Hence, this study elucidates the molecular mechanism of aescin in targeting protein kinases and suggests that it could be a safer and more viable therapeutic agent for lung cancer treatment.


Assuntos
Neoplasias Pulmonares , Saponinas , Triterpenos , Humanos , Escina/farmacologia , Escina/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Saponinas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Triterpenos/farmacologia , Linhagem Celular Tumoral , Apoptose , Serina-Treonina Quinases TOR/metabolismo , Pulmão/metabolismo , Autofagia
8.
Life Sci ; 309: 120964, 2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36115584

RESUMO

INTRODUCTION AND AIM: Purpurin, a naturally occurring anthraquinone isolated from the roots of Rubia cordifolia, exhibits anti-cancer, anti-genotoxic, anti-microbial, neuromodulatory and photodynamic activity. However, purpurin's in vivo and in vitro antioxidant mechanism remains unexplored. The present study explores the anti-oxidative mechanism of purpurin under the influence of alcohol using in vivo and in vitro test systems. METHODS: Mice hepatocytes and alcohol-induced liver toxicity model were used to evaluate the effect of purpurin. The non-enzymatic and enzymatic oxidative stress markers were estimated by the colorimetric method. The reactive oxygen species (ROS) were quantified in mitochondria and cells using flow cytometer. Real-time PCR and western blotting were used to quantify cytochrome 450 subtype 2E1 (CYP2E1) and Nrf2 expression in the liver tissue of mice. In silico studies were performed through receptor-ligand binding interaction. KEY FINDINGS: Purpurin effectively reduced total cellular and mitochondrial ROS in primary hepatocytes and WRL-68 cells. It prevented alcohol-induced ROS-dependent biochemical and cellular insults observed by analysing the serum glutamic pyruvic transaminase (SGPT), glutamic-oxaloacetic transaminase (SGOT) levels and CYP2E1 expression in liver tissue of alcohol-administered mice. Moreover, it also restored the activity of antioxidant enzymes. Its antioxidant effect was established by glutathione and ROS-dependent mechanisms using buthionine sulfoximine and N-acetyl cysteine. Along with alcohol, purpurin up-regulated Nrf2 expression in hepatocytes. SIGNIFICANCE: This work confirmed the ameliorative effect of purpurin for alcohol-induced hepatotoxicity by drabbing free radicals and curbing oxidative stress via activation of antioxidant signalling pathways.


Assuntos
Antraquinonas , Doença Hepática Induzida por Substâncias e Drogas , Etanol , Fator 2 Relacionado a NF-E2 , Animais , Camundongos , Alanina Transaminase/metabolismo , Antraquinonas/farmacologia , Antioxidantes/farmacologia , Aspartato Aminotransferases/metabolismo , Butionina Sulfoximina/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Cisteína/farmacologia , Citocromo P-450 CYP2E1/metabolismo , Etanol/toxicidade , Glutationa/metabolismo , Ligantes , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
9.
Front Cell Infect Microbiol ; 12: 836819, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35909977

RESUMO

The inhibition/degradation potential of Carissa carandas proteinaceous leaf extract against mixed bacterial biofilm of Staphylococcus aureus MTCC 96, Escherichia coli MTCC 1304, Pseudomonas aeruginosa MTCC 741, and Klebsiella pneumoniae MTCC 109, responsible for nosocomial infections, was evaluated. Distinct inhibition/degradation of mixed bacterial biofilm by the proteinaceous leaf extract of C. carandas was observed under a microscope, and it was found to be 80%. For mono-species biofilm, the maximum degradation of 70% was observed against S. aureus biofilm. The efficiency of aqueous plant extracts to inhibit the mono-species biofilm was observed in terms of minimum inhibitory concentration (MIC), and the best was found against P. aeruginosa (12.5 µg/ml). The presence of flavonoids, phenols, and tannins in the phytochemical analysis of the plant extract suggests the main reason for the antibiofilm property of C. carandas. From the aqueous extract, protein fraction was precipitated using 70% ammonium sulfate and dialyzed. This fraction was purified by ion-exchange chromatography and found to be stable and active at 10°C (pH 7). The purified fraction showed less than 40% cytotoxicity, which suggests that it can be explored for therapeutic purposes after in-depth testing. In order to investigate the mechanistic action of the biofilm inhibition, the plant protein was tested against Chromobacterium violaceum CV026, and its inhibitory effect confirmed its quorum quenching nature. Based on these experimental analyses, it can be speculated that the isolated plant protein might influence the signaling molecule that leads to the inhibition effect of the mixed bacterial biofilm. Further experimental studies are warranted to validate our current findings.


Assuntos
Apocynaceae , Percepção de Quorum , Antibacterianos/química , Bactérias , Biofilmes , Extratos Vegetais , Proteínas de Plantas/farmacologia , Pseudomonas aeruginosa , Staphylococcus aureus , Virulência
10.
J Biomol Struct Dyn ; 39(9): 3312-3323, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32362196

RESUMO

Cirsimaritin is a dimethoxy flavone, which is present in Ocimum sanctum, Microtea debilis, Artemisia judaica, Cirsium japonicum, and Lithocarpus dealbatus. Its antiproliferative potential has been explored in breast and gall bladder cancer cell lines. However, no reports are available on skin and squamous lung carcinoma. Also, the complete mode of action is unknown. Therefore, in the present study, the anticancer potential of cirsimaritin is explored in organ-specific cell lines by using MTT assay. Further, the inhibitory potential and binding interaction with the selected targets were analyzed through in vitro and in-silico analysis. Cirsimaritin showed selective anticancer activity against NCIH-520 cell-line (IC50 23.29 µM), also inhibited the proliferation of other cell-lines up to 48% at 100 µM. In NCIH-520 cell-line, cirsimaritin significantly increased the apoptosis of the cells at both the tested concentrations (10 and 100 µM), which was confirmed by Annexin-V signifying the induction of late apoptosis. Besides, an increase in the ROS levels of 1.6 fold (10 µM) and 1.8 fold (100 µM), circimaritin also inhibits the activity of ODC and CATD with the IC50 57.30 and 68.22 µM respectively. It exhibited a good binding score with the selected targets, follow Lipinski's rule of five and non-mutagenic. Hence, cirsimaritin is a potent molecule, which inhibits the proliferation of lung squamous cell lines by inducing apoptosis. It also inhibited the activity of ODC and CATD responsible for the progression phase in the cancer cells. Communicated by Ramaswamy H. Sarma.


Assuntos
Carcinoma de Células Escamosas , Flavonas , Neoplasias Pulmonares , Apoptose , Carcinoma de Células Escamosas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células , Flavonas/farmacologia , Humanos , Pulmão , Neoplasias Pulmonares/tratamento farmacológico
11.
Curr Top Med Chem ; 20(20): 1791-1809, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32357817

RESUMO

BACKGROUND: In chemotherapy for cancer, conventional drugs aim to target the rapidly growing and dividing cells at the early stages. However, at an advanced stage, cancer cells become less susceptible because of the multidrug resistance and the recruitment of alternative salvage pathways for their survival. Besides, owing to target non-selectivity, healthy proliferating cells also become vulnerable to the damage. The combination therapies offered using flavonoids to cure cancer not only exert an additive effect against cancer cells by targetting supplementary cell carnage pathways but also hampers the drug resistance mechanisms. Thus, the review aims to discuss the potential and pharmacokinetic limitations of flavonoids in cancer treatment. Further successful synergistic studies reported using flavonoids to treat cancer has been described along with potential drug delivery systems. METHODS: A literature search was done by exploring various online databases like Pubmed, Scopus, and Google Scholar with the specific keywords like "Anticancer drugs", "flavonoids", "oncology research", and "pharmacokinetics". RESULTS: Dietary phytochemicals, mainly flavonoids, hinder cell signalling responsible for multidrug resistance and cancer progression, primarily targeting cancer cells sparing normal cells. Such properties establish flavonoids as a potential candidate for synergistic therapy. However, due to low absorption and high metabolism rates, the bioavailability of flavonoids becomes a challenge. Such challenges may be overcome using novel approaches like derivatization, and single or co-delivery nano-complexes of flavonoids with conventional drugs. These new approaches may improve the pharmacokinetic and pharmacodynamic of flavonoids. CONCLUSION: This review highlights the application of flavonoids as a potential anticancer phytochemical class in combination with known anti-cancer drugs/nanoparticles. It also discusses flavonoid's pharmacokinetics and pharmacodynamics issues and ways to overcome such issues. Moreover, it covers successful methodologies employed to establish flavonoids as a safe and effective phytochemical class for cancer treatment.


Assuntos
Antineoplásicos/uso terapêutico , Flavonoides/uso terapêutico , Neoplasias/tratamento farmacológico , Antineoplásicos/química , Antineoplásicos/farmacocinética , Disponibilidade Biológica , Sistemas de Liberação de Medicamentos , Flavonoides/química , Flavonoides/farmacocinética , Neoplasias/metabolismo
12.
Curr Top Med Chem ; 20(21): 1898-1909, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32648844

RESUMO

BACKGROUND: Since centuries plant-based compounds are known for the treatment of cancer in both traditional and contemporary medicine. The problems like target non-specificity and toxicity are well-known regarding anticancer drugs. Therefore, target specific search of novel entities is constant. Isothymusin is a dimethoxy, trihydroxy flavone present in plants like Ocimum sanctum, and Limnophilla geoffrayi. There are limited reports available on the anticancer potential of isothymusin. OBJECTIVES: The effects of isothymusin on redox status, cell cytotoxicity, and targets involved in the promotion and progression of the cancer cells have been investigated. METHODS: Antiproliferative efficacy was evaluated by MTT, Neutral Red Uptake, and Sulforhodamine-B assays. The spectrophotometric methods were adopted to study the effect against selected targets. Redox activity was assessed by in vitro antioxidant assays and the interaction study, ADMET profiling, and toxicity assessments were done in silico. RESULTS: Isothymusin scavenges the radicals, i.e., DPPH and nitric oxide with moderate ferric reducing potential. It affected the proliferation of leukemia, colon, skin, and breast cancer cell lines by more than 50% but moderately affected prostate, kidney, lung, hepatic, and breast adenocarcinoma (up to 48%). Isothymusin inhibited the enzymes associated with the promotion stage of cancer, including cycloxygenase- 2 and lipoxygenase-5. Additionally, it also inhibited the activity of proliferation markers like cathepsin- D, dihydrofolate reductase, hyaluronidase, and ornithine-decarboxylase. Besides, in silico studies supported the in vitro enzyme inhibition assays outcome. Toxicity studies showed promising results of chemical descriptors and non-skin-irritant, moderate ocular-irritancy, and in vitro Ames test confirmed non-mutagenic nature. CONCLUSION: Isothymusin showed radical scavenging and anti-proliferative activities, which may be taken up as a phytochemical lead for the synthesis of analogues possessing enhanced anticancer potential.


Assuntos
Antineoplásicos/farmacologia , Antioxidantes/farmacologia , Flavonoides/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Antioxidantes/síntese química , Antioxidantes/química , Compostos de Bifenilo/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Flavonoides/síntese química , Flavonoides/química , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Picratos/antagonistas & inibidores , Relação Estrutura-Atividade
13.
Food Chem Toxicol ; 143: 111550, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32640357

RESUMO

Cirsilineol belonging to the flavones category have not been explored in detail for anti-proliferative potential, therefore selected for the investigation. Hence, the antiproliferative potential of cirsilineol has been established in NCIH-520 cells. Cirsilineol exhibited good binding-energy and inhibited the activity of ODC, CATD, DHFR, HYAL, LOX-5, and COX-2 up to 45.14% at 100 µM. It significantly inhibited the proliferation of NCIH-520 cells (81.96%) and likewise, the proliferation of other cell lines up to 48.50%. It also induced an increase in the sub-diploid cell population, which then leads to an increase in apoptosis by 2.64 and 5.12 fold at 10 µM and 100 µM respectively. Further, the Annexin-V-FITC assay confirmed the late apoptosis and necrosis in the NCIH-520 cell line induced by cirsilineol. The ROS production was enhanced by 1.16 and 2.22 folds at 10 µM and 100 µM respectively. Besides, cirsilineol revealed acceptable ADME properties, non-toxic and non-mutagenic compound. Altogether, these findings provide evidence that cirsilineol inhibited the proliferation of NCIH-520 cells by inducing ROS-mediated apoptosis and offer new insight into the anti-proliferative potential of cirsilineol, which can further be exploited to either synthesise new derivatives or its candid usage as a herbal lead for cancer treatment.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Flavonas/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Humanos , Estrutura Molecular , Espécies Reativas de Oxigênio
14.
Sci Rep ; 10(1): 9102, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32499562

RESUMO

We determined the role of cellular fibronectin (CFN) containing the alternatively spliced extra domain A (FN-EDA) in causing insulin resistance (IR) through toll-like receptor 4 (TLR4). Circulating FN-EDA level was evaluated in mouse and rat IR models. Specific anti-FN-EDA antibody and TLR4 inhibitor were used to study its role in IR in mice. CFN protein was injected to evaluate TLR4 dependent effect of FN-EDA in IR. Furthermore, FN-EDA was estimated in blood plasma and correlated with demographic and clinical characteristics in healthy human participants (n = 38). High-fat diet feeding significantly increased circulating FN-EDA in both mouse (P = 0.03) and rat (P = 0.02) IR models. Antibody against FN-EDA protected mice from IR by increasing glucose disposal rate following glucose (P = 0.02) and insulin (P = 0.01) tolerance tests. CFN protein injection caused IR, however, TLR4 inhibitor protected the mice from CFN induced IR. Multivariate regression analysis predicted an independent positive correlation between circulating FN-EDA and fasting plasma glucose (P = 0.003) in healthy human participants. In conclusion, FN-EDA may cause IR through TLR4 by decreasing glucose disposal rate following glucose and insulin load. Targeting FN-EDA thus can be considered as a possible therapeutic strategy to delay prediabetes progression to diabetes.


Assuntos
Diabetes Mellitus/etiologia , Fibronectinas/efeitos adversos , Fibronectinas/fisiologia , Resistência à Insulina/genética , Receptor 4 Toll-Like/metabolismo , Animais , Glicemia/metabolismo , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/prevenção & controle , Modelos Animais de Doenças , Feminino , Fibronectinas/metabolismo , Voluntários Saudáveis , Humanos , Masculino , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular , Ratos , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Receptor 4 Toll-Like/antagonistas & inibidores
15.
Injury ; 51(7): 1448-1456, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32430194

RESUMO

INTRODUCTION: Tibial plafond fractures (TPF) are complex injuries often resulting in poor outcomes. Combination of articular impaction, metaphysealcomminution and soft-tissue injury results in a significant treatment challenge. The aim of this study was to conduct a systematic review and meta-analysis to compare post-operative complications and functional outcomes of open reduction and internal fixation (ORIF) versus circular external fixation (CEF) for treatment of TPF. METHODS: A comprehensive search of PubMed/MEDLINE, Embase, Scopus and Cochrane library was undertaken. All studies published in English language comparing ORIF with CEF for treatment of TPF were included. RESULTS: 5 comparative studies with 239 fractures met the inclusion criteria. Meta-analysis showed no significant difference in rates of non-union, malunion, superficial infection, deep infection, and secondary arthrodesis between the two treatment groups. Significantly higher rate of unplanned metalwork removal (RR 5.68, 95% CI 1.13 to 28.55, p = 0.04) and lower rate of post-traumatic arthritis (RR 0.48, 95% CI 0.30 to 0.78, p = 0.003) were found in patients that underwent ORIF. 1 study showed significantly lower functional outcomes scores with CEF (p< 0.05), whereas 3 studies found comparable functional outcomes between the two treatment groups. Overall, there was a preference in treating more severe injuries with CEF. CONCLUSION: CEF and ORIF are both acceptable treatment options for surgical management of TPF, with comparable post-operative complication rates and functional outcomes. This study highlights paucity of high-quality evidence regarding the optimal fixation method for TPF.


Assuntos
Fixadores Externos , Fixação Interna de Fraturas , Redução Aberta , Fraturas da Tíbia/cirurgia , Consolidação da Fratura , Humanos , Fraturas Intra-Articulares/cirurgia , Desempenho Físico Funcional , Complicações Pós-Operatórias
16.
Pathogens ; 9(12)2020 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-33276564

RESUMO

Tick bites in Australia are linked to the transmission of a variety of infectious diseases in humans, livestock and wildlife. Despite this recognition, little is currently known about the variety of potential pathogens that are carried and transmitted by Australian ticks. In this study, we attempted to expand knowledge of Australian tick-borne bacterial pathogens by analyzing various tick species from the state of Queensland for potential human pathogens belonging to the Rickettsia, Coxiella and Borrelia genera. A total of 203 ticks, comprising of four genera and nine different tick species, were screened by specific qPCR assays. An overall Rickettsia qPCR positivity of 6.4% (13/203) was detected with rickettsial DNA found in four tick species (Ixodes holocyclus, I. tasmani, Amblyommatriguttatum, and Haemaphysalis longicornis). Amplification and analysis of several rickettsial genes from rickettsial qPCR positive samples identified sequences closely related to but genetically distinct from several previously described cultured and uncultured rickettsial species in the Rickettsia spotted fever subgroup. No ticks were positive for either Coxiella or Borrelia DNA. This work suggests that a further diversity of rickettsiae remain to be described in Australian ticks with the full importance of these bacteria to human and animal health yet to be elucidated.

17.
Behav Brain Res ; 361: 113-121, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30584898

RESUMO

Excessive consumption of high-fat fructose diet (HFFD) is associated with the development of systemic insulin resistance (InsRes) and further progression into type-2 diabetes (T2DM). InsRes induced hippocampal insulin signaling has serious consequence on hampered sensorimotor, cognitive performance and long term potentiation accompained to neuronal cell death in hippocampus. However, short-term HFFD/Streptozotocin (STZ) mediated hippocampal InsRes and related neurobehavioral alterations in adoloscents have not been reported. Therefore, we investigated a one-week HFFD model to augment the state of InsRes along with a single sub-diabetogenic dose of STZ (45 mg/kg i.p) to produce a hampered hippocampal insulin signaling associated with frank hyperglycemia and other biochemical and neurobehavioral alterations in young rats. To achieve this, male wistar rats of age (8-10 weeks) and weight 80-120 g were divided into two main groups: (1) fed with commercial standard normal fat diet (NFD: 6.5% kcal fat) and (2) fed an in-house prepared high-fat diet [HFFD: 58% kcal fat] and 20% high-fructose corn syrup in the distilled water. Our results showed an increase in calorie intake, water intake, body weight and blood glucose levels. Further, an increase in fasting serum insulin and Homeostasis Model Assessment-index (HOMA-I) and oral glucose tolerance test (OGTT) was observed. Whereas, we observed a decrease in hippocampal insulin signaling and translocation of glucose transporter type 4 (GLUT4) to neuronal membrane. Further, HFFD/STZ mediated oxidative stress, lipid peroxidation (LPO), decreased antioxidant levels, Brain-derived neurotrophic factor (BDNF) levels and further activation of increase caspase-3 was observed. These battery of events indicate biochemical alterations in hippocampus resulting in cognition and memory deficit.


Assuntos
Dieta Ocidental/efeitos adversos , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Peso Corporal , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cognição/efeitos dos fármacos , Dieta Hiperlipídica , Glucose/metabolismo , Hipocampo/metabolismo , Homeostase/efeitos dos fármacos , Hiperglicemia/metabolismo , Insulina/metabolismo , Resistência à Insulina/fisiologia , Masculino , Memória/efeitos dos fármacos , Transtornos da Memória/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
18.
Pharmacol Biochem Behav ; 169: 35-47, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29654794

RESUMO

Binge eating disorder (BED) is a stress-related disorder characterized by acute episodes of excessive food intake. Piracetam, a nootropic agent has been reported to show several other neuropharmacological properties. The present study, evaluated the pharmacological effect of piracetam (200 mg/kg i.p.) on BED in female rats, induced by free access to palatable cookies for 2 h on alternate days. BED was confirmed by an increase in binge eating behavior and weight gain. BED leads to anxiety, cognitive and memory deficits, as evaluated by EPM (Elevated plus maze), OFT (open field test), and Y-maze tests. Increased levels of plasma corticosterone (CORT), glutamate in nucleus accumbens (NAC), hypothalamus (HYP) and prefrontal cortex (PFC) indicate stress and excitotoxicity. Moreover, it was observed that the levels of dopamine were higher in NAC and PFC, and less in HYP which may be responsible for motivational behavior for palatable feeding and cognitive deficits. More surprisingly, feeding behaviour regulating hormones namelyleptin was increased and ghrelin level was decreased in BED. Further, level of acetylcholine which regulates cognitive behaviour was compromised in BED. Piracetam significantly decreased binge eating behavior and associated body weight and regulated the levels of concerned neurotransmitters in respective regions. However, piracetam did not alter normal feeding behavior in the fast-refed model. Further, piracetam showed brain region-specific decrease in vascular endothelial growth factor expression. Piracetam showed anxiolytic activity and also alleviated cognitive deficit observed in BED. Hence, preclinical evidence indicates the potential use of piracetam for the treatment of BED.


Assuntos
Bulimia/prevenção & controle , Nootrópicos/farmacologia , Piracetam/farmacologia , Acetilcolina/metabolismo , Animais , Bulimia/metabolismo , Transtornos Cognitivos/prevenção & controle , Corticosterona/sangue , Dopamina/sangue , Comportamento Alimentar/efeitos dos fármacos , Feminino , Hipotálamo/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Neurotransmissores/metabolismo , Núcleo Accumbens/metabolismo , Córtex Pré-Frontal/metabolismo , Ratos Wistar , Aumento de Peso/efeitos dos fármacos
19.
ChemMedChem ; 13(4): 384-395, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29319226

RESUMO

Muscarinic acetylcholine receptors (mAChRs) are important therapeutic targets for several diseases of the central nervous system and periphery. However, the lack of subtype-selective ligands for these receptors is a major challenge. A novel approach involving the integration of a natural product framework with a bioactive molecule (iNPBM) by using gephyrotoxin and the isoindoline framework is demonstrated for the discovery of new and selective mAChR modulators. We established a scalable and versatile synthetic scheme to enable the synthesis of various analogues that provided the first structure-activity relationship study of this class of compounds. Pharmacological profiling of these compounds demonstrated several ligands with high affinity and selectivity for mAChRs. Specifically, RG-06 and RG-09 were found to be antagonists of M3-mAChR, whereas RG-02 was found to be an agonist at M2-mAChR. Furthermore, RG-02 exhibited salutary effects in an established pharmacological model of a cognitive deficit in mice.


Assuntos
Produtos Biológicos/metabolismo , Receptores Muscarínicos/metabolismo , Alcaloides/química , Alcaloides/metabolismo , Alcaloides/farmacologia , Animais , Produtos Biológicos/química , Produtos Biológicos/farmacologia , Catálise , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Isoindóis/química , Isoindóis/metabolismo , Isoindóis/farmacologia , Locomoção/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Metais/química , Camundongos , Camundongos Endogâmicos C57BL , Conformação Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Receptores Muscarínicos/química , Relação Estrutura-Atividade
20.
J Thorac Dis ; 9(8): 2703-2713, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28932579

RESUMO

BACKGROUND: Lung cancer is increasingly a disease of the elderly and frail population with a median age of 70 years at diagnosis. Therefore, consideration of the impact of interventions on health-related quality of life (HRQOL) and not only absolute survival is especially important. For non-small cell lung cancer (NSCLC), video-assisted thoracoscopic surgery (VATS) has been gaining popularity over the last few decades, replacing traditional open lobectomies. For high-risk patients who are not deemed suitable for surgery, stereotactic ablative body radiotherapy (SABR) provides a potentially curative alternative. However, little is known about how VATS and SABR affect HRQOL measured using patient reported outcome measures (PROMs). The LiLAC study (Life after Lung Cancer) aims to explore HRQOL following intervention with VATS or SABR using validated PROMs and to pilot the use of an online questionnaire system (QTool) in this setting. We hope the results will aid both patients and clinicians in decision making and improve the management of post-intervention problems. METHODS: In total, 300 patients (150 VATS and 150 SABR) patients will be recruited over the study period. Patients will be approached prior to intervention and asked to complete baseline HRQOL questionnaires. They will be given access to the QTool online system and then in the 12 months following intervention will be asked to complete questionnaires (paper or online) at 4-time points. Answers will available for patients and clinicians to view throughout the study period. Clinical information (age, gender, co-morbidity, current medications and smoking status along with treatment-specific information) will also be collected. Primary outcome will be to detect changes of PROs (HRQOL and patient satisfaction) after VATS lung resections or SABR in early stage lung cancer patients. Secondary outcomes include correlation of patient's clinical data with HRQOL results to identify predictors of poor outcomes and exploration of patient and clinician views on the usefulness of QOL measurements. DISCUSSION: (I) This first study will primarily compare multiple patients reported outcomes for 12 months after VATS lobectomy and SABR in early stages NSCLC patients. We will explore the acceptability of an online assessment of the HRQOL in NSCLC patients. (II) The study is also focused on the patients' opinion during the shared decision-making process, which has rarely been investigated in surgical lung cancer patients. (III) This is not a randomised trial. As a consequence, inherent cohort selection bias and unknown or unaccounted confounders correlated with the outcome of interest may influence the results of the comparison between the treatment groups. (IV) LILAC is not looking at a direct comparison, but to depict the trajectory of recovery post-treatments and preservation or improvement of the HRQOL. This study has received ethical approval from NRES Yorkshire and the Humber- Leeds East Research Ethics Committee (REC Ref: 16/YH/0407). Results of this study will be shared with participating hospitals and made available to the academic community through submission for publication in international peer-reviewed journals and presentation at relevant national and international conferences. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02882750.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA