Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cancer Res Ther ; 18(3): 668-676, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35900539

RESUMO

Background: Dysregulation in Wnt/ß-catenin signaling has been associated with the initiation and metastasis of cancer cells. Transcription factor 4 (TCF4) (also named as transcription factor 7-like 2) is a key transcriptional factor of the Wnt signaling pathway, which, when interact with ß-catenin activates Wnt genes which plays an essential role in tumor development. The expression pattern and clinical significance of TCF4 in gallbladder cancer (GBC) are not yet established. Aims: This study was performed to assess the expression pattern of TCF4 in GBC tissue and attempted to correlate its expression with different clinicopathological parameters. Materials and Methods: The study was conducted on 33 surgically resected specimens of gallbladder carcinoma (GBC) and 12 cases of chronic cholecystitis (CC) as control, which had been confirmed from histology. The expression of TCF4 was performed by the reverse transcription polymerase chain reaction and immunohistochemistry. Results: Relative mRNA expression levels of ß-catenin and TCF4 in GBC tissues were significantly (P < 0.05) higher than in CC samples. TCF4 protein expression was observed in 81.82% (27/33) GBC cases. Specifically, among GBC samples, 21.21% (7/33) was graded as strongly positive, 60.61% (20/33) graded as moderately positive, whereas 18.18% (6/33) graded as negative. All 12 CC samples graded as negative. Overall, TCF4 expression in GBC tissues was statistically significant over CC samples (P < 0.05). Moreover, we observed that TCF4 expression was significantly higher (P < 0.05) in high tumor grades than low grade, higher (P < 0.05) in Stage 2 and Stage 3 than Stage 1. Conclusion: The present study suggests that TCF4 may exert an oncogenic role in the progression of GBC and may serve as a new potential candidate biomarker for tumor progression, and it might be a potential therapeutic target against GBC.


Assuntos
Colecistite , Neoplasias da Vesícula Biliar , Fator de Transcrição 4/metabolismo , Linhagem Celular Tumoral , Colecistite/patologia , Neoplasias da Vesícula Biliar/patologia , Humanos , Fator de Transcrição 4/genética , Fatores de Transcrição/genética , Via de Sinalização Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
2.
Eur J Med Chem ; 198: 112257, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32375073

RESUMO

In our overall goal to overcome the limitations associated with natural products for the management of Alzheimer's disease and to develop in-vivo active multifunctional cholinergic inhibitors, we embarked on the development of ferulic acid analogs. A systematic SAR study to improve upon the cholinesterase inhibition of ferulic acid with analogs that also had lower logP was carried out. Enzyme inhibition and kinetic studies identified compound 7a as a lead molecule with preferential acetylcholinesterase inhibition (AChE IC50 = 5.74 ± 0.13 µM; BChE IC50 = 14.05 ± 0.10 µM) compared to the parent molecule ferulic acid (% inhibition of AChE and BChE at 20 µM, 15.19 ± 0.59 and 19.73 ± 0.91, respectively). Molecular docking and dynamics studies revealed that 7a fits well into the active sites of AChE and BChE, forming stable and strong interactions with key residues Asp74, Trp286, and Tyr337 in AChE and with Tyr128, Trp231, Leu286, Ala328, Phe329, and Tyr341 in BChE. Compound 7a was found to be an efficacious antioxidant in a DPPH assay (IC50 = 57.35 ± 0.27 µM), and it also was able to chelate iron. Data from atomic force microscopy images demonstrated that 7a was able to modulate aggregation of amyloid ß1-42. Upon oral administration, 7a exhibited promising in-vivo activity in the scopolamine-induced AD animal model and was able to improve spatial memory in cognitive deficit mice in the Y-maze model. Analog 7a could effectively reverse the increased levels of AChE and BChE in scopolamine-treated animals and exhibited potent ex-vivo antioxidant properties. These findings suggest that 7a can act as a lead molecule for the development of naturally-inspired multifunctional molecules for the management of Alzheimer's and other neurodegenerative disorders.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Produtos Biológicos/química , Colinérgicos/química , Inibidores da Colinesterase/química , Colinesterases/metabolismo , Ácidos Cumáricos/química , Sequência de Aminoácidos , Peptídeos beta-Amiloides/metabolismo , Animais , Antioxidantes/química , Antioxidantes/farmacologia , Produtos Biológicos/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Colinérgicos/farmacologia , Inibidores da Colinesterase/farmacologia , Disfunção Cognitiva/tratamento farmacológico , Ácidos Cumáricos/farmacologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Modelos Moleculares , Fragmentos de Peptídeos/química , Escopolamina/metabolismo , Memória Espacial/efeitos dos fármacos
3.
Int Immunopharmacol ; 77: 106002, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31711939

RESUMO

Antibodies targeting PD1 receptor have emerged as a promising therapeutic strategy against multiple types of solid cancers. However, relatively low complete response rates observed with anti-PD1 mAb monotherapy emphasizes the importance of testing new immunotherapeutic combinations. The production of extracellular adenosine in solid tumors was recently identified as a major immunosuppressive pathway, targeting this pathway would enhance the therapeutic activity of anti-PD1 mAbs. In this study, we evaluated the anti-tumor activity and mechanism of action of caffeine and anti-PD1 mAb combination therapy against carcinogen- and cell line-induced tumors. Our results demonstrate that combination therapy enhanced the anti-tumor activity and prolonged overall survival period against 3-MCA-induced tumors. In addition, combination therapy showed a significant anti-tumor activity against B16F10 melanoma tumors. We found that combination therapy showed additive increase in infiltration of CD4+ and CD8+ T lymphocytes into the B16F10 melanoma tumors. On the other hand, combination therapy showed significant decrease in infiltration of CD4+CD25+ T regulatory cells. We further investigated whether the observed anti-tumor effect of caffeine and anti-PD1 mAb combination therapy is mediated through the release of cytokines. We found that caffeine and anti-PD1 mAb combination therapy significantly increased intra-tumoral TNF-α and IFN-γ levels. Our work suggests that administration of caffeine and anti-PD1 mAb harness the therapeutic potential of effector T cells in vivo possibly due to combined blockade of PD1 and adenosine-A2A receptor pathway. This study provides the scientific basis for testing combination regimens of caffeine and anti-PD1 mAbs for sustained tumor control in cancer patients.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Cafeína/farmacologia , Melanoma/tratamento farmacológico , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Terapia Combinada/métodos , Feminino , Imunossupressores/farmacologia , Imunoterapia/métodos , Interferon gama/metabolismo , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo
4.
Biomed Pharmacother ; 105: 312-319, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29864619

RESUMO

Caffeine (1,3,7-trimethylxanthine) is one of the most commonly consumed food ingredient throughout the world for thousands of years. It is naturally present in beans, leaves, seeds, and fruits of more than 60 plants and the most common sources are cocoa beans, coffee beans, tea leaves, kola nuts, and guarana berries. Several epidemiological studies suggested that consumption of coffee reduces the risk of different types of cancers. Understanding the chemotherapeutic molecular mechanisms of caffeine would facilitate in designing efficacious combination strategies with other anticancer drugs. Therefore, the aim of this review is to identify and highlight all the mechanisms involved in chemotherapeutic activity of caffeine. Mechanistically, caffeine has been shown to affect cell cycle progression at checkpoints, induce apoptosis, and inhibit drug efflux from cells. Caffeine at lower (micromolar) and relatively non-toxic concentrations has been shown to promote anti-tumor immune response (A2 A) and to inhibit tumor angiogenesis (A3) and migration (A2B) by antagonizing adenosine receptors. Considering the fact that a lower (micromolar) and relatively non-toxic concentrations of caffeine are required for its anti-tumor immune response and antiangiogenic activity, developing a combination strategy with immune checkpoint (PD-1 or CTLA-4) inhibitors would represent a novel approach to treat cancer.


Assuntos
Antineoplásicos/uso terapêutico , Cafeína/uso terapêutico , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Cafeína/farmacologia , Humanos , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/patologia
5.
Brain Res ; 1674: 1-9, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28827076

RESUMO

Diabetes mellitus and depression are the common comorbid disorders affecting humans worldwide. There is an unmet need to develop therapeutic strategies to treat both diabetes mellitus and comorbid depression. The present study evaluated the effectiveness of metformin and ascorbic acid against type 2 diabetes mellitus and comorbid depression in rats. Four groups of diabetic rats were orally administered with vehicle (1mL/kg), metformin (25mg/kg), ascorbic acid (25mg/kg), or combination of metformin (25mg/kg) and ascorbic acid (25mg/kg) for 11 consecutive days. Diabetes was induced by single-dose administration of streptozotocin (65mg/kg, i.p.) with nicotinamide (120mg/kg, i.p.). Comorbid depression was induced by five inescapable foot-shocks (2mA, 2ms duration) at 10s intervals on days 1, 5, 7, and 10. One group of healthy rats received only vehicles to serve as nondiabetic control group. On day 11, animals were sacrificed, and blood and brain samples were collected from each rat following forced swim test. Plasma glucose, insulin, and corticosterone levels were estimated in plasma. The levels of monoamines, proinflammatory cytokines, and oxidative stress were measured in prefrontal cortex. The combination therapy significantly reduced immobility period, glucose, and corticosterone levels relative to diabetes with comorbid depression group. Furthermore, the combination therapy increased the levels of insulin and monoamines, and caused a significant reductions in oxidative stress and proinflammatory cytokines. In conclusion, the present study revealed that metformin and ascorbic acid combination therapy could be a potential strategy to treat type 2 diabetes mellitus and comorbid depression.


Assuntos
Ácido Ascórbico/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Metformina/farmacologia , Animais , Ácido Ascórbico/metabolismo , Monoaminas Biogênicas/metabolismo , Glicemia , Comorbidade , Corticosterona/sangue , Citocinas/efeitos dos fármacos , Depressão/tratamento farmacológico , Transtorno Depressivo/tratamento farmacológico , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/complicações , Modelos Animais de Doenças , Quimioterapia Combinada , Insulina/sangue , Masculino , Metformina/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Endogâmicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...